3,892
Views
23
CrossRef citations to date
0
Altmetric
Review Article

Nutraceutical approaches to metabolic syndrome

, , & ORCID Icon
Pages 678-697 | Received 10 Jul 2017, Accepted 07 Aug 2017, Published online: 22 Aug 2017

Abstract

Metabolic Syndrome (MetS), affecting at least 30% of adults in the Western World, is characterized by three out of five variables, from high triglycerides, to elevated waist circumference and blood pressure. MetS is not characterized by elevated cholesterolemia, but is rather the consequence of a complex interaction of factors generally leading to increased insulin resistance. Drug treatments are of difficult handling, whereas well-characterized nutraceuticals may offer an effective alternative. Among these, functional foods, e.g. plant proteins, have been shown to improve insulin resistance and reduce triglyceride secretion. Pro- and pre-biotics, that are able to modify intestinal microbiome, reduce absorption of specific nutrients and improve the metabolic handling of energy-rich foods. Finally, specific nutraceuticals have proven to be of benefit, in particular, red-yeast rice, berberine, curcumin as well as vitamin D. All these can improve lipid handling by the liver as well as ameliorate insulin resistance. While lifestyle approaches, such as with the Mediterranean diet, may prove to be too complex for the single patient, better knowledge of selected nutraceuticals and more appropriate formulations leading to improved bioavailability will certainly widen the use of these agents, already in large use for the management of these very frequent patient groups.

    Key messages

  • Functional foods, e.g. plant proteins, improve insulin resistance.

  • Pro- and pre-biotics improve the metabolic handling of energy-rich foods.

  • Nutraceutical can offer a significant help in handling MetS patients being part of lifestyle recommendations.

Introduction

Metabolic syndrome (MetS) is present as a common clinical condition in countries where obesity and Western unhealthy dietary patterns prevail. The incidence of MetS is increasing to epidemic proportions thus entailing substantial health care costs. Indeed, MetS associates with an elevated risk of cardiovascular disease and type 2 diabetes (T2D), as well as other conditions, such as non-alcoholic fatty liver disease.

After the first formal definition of MetS introduced in 1998 by the World Health Organization (WHO), several expert panels have attempted to introduce diagnostic criteria. In 2001 the National Cholesterol Education Program (NCEP) Adult Treatment Panel III (ATP III) [Citation1] recognized for the first time the clusterization of the metabolic risk factors, these being also cardiovascular risk factors. In 2003, the American Association of Clinical Endocrinologists modified the ATP III criteria highlighting the central role of insulin resistance in the pathogenesis of the syndrome [Citation2]. In 2005, the International Diabetes Federation (IDF) issued a consensus document aimed at producing a clinically useful definition of Mets to identify individuals at high risk of cardiovascular disease and T2D on a worldwide basis [Citation3]. In the same year, the American Heart Association (AHA)/National Heart, Lung and Blood Institute (NHLBI) suggested criteria for the diagnosis of MetS [Citation3]. Finally, IDF, NHBLI, AHA, World Heart Federation and the International Association for the Study of Obesity produced a joint statement called the “Harmonization definition”, which, as of today, represents the most common recognized criteria for the clinical diagnosis of MetS [Citation4], summarized in .

Table 1. Risk factors for the clinical diagnosis of metabolic syndrome.

The present review will address the management of MetS by the use of specific nutraceuticals, in particularly plant proteins, pro/prebiotics, curcumin, berberine, red yeast rice and vitamin D. The activity of these substances will be analyzed based on the described major MetS features. While in many cases, e.g. MetS linked to elevated blood pressure, drug treatments may be of higher efficacy, the present favourable view of nutraceuticals by patients and by the medical community, makes it mandatory to carefully analyze what these products may offer in terms of improvement of metabolic health in MetS. The major features of MetS will be hereafter outlined.

Metabolic syndrome: a deep overview

Body variables

Abdominal obesity is a prominent feature of MetS contributing to an elevation of cardiovascular risk. Indeed, excess abdominal adipose tissue releases large amounts of FFAs via the portal system to the liver, and may interfere with hepatic insulin clearance [Citation5], thus contributing to IR [Citation6]. In addition, it is an active endocrine organ, secreting different adipokines, including leptin, adiponectin, resistin, interleukins (IL), and tumour necrosis factor (TNF)-α [Citation7]. In the case of excess abdominal weight, there is evidence of macrophage infiltration into the adipose tissue [Citation8], contributing to the inflammatory profile reported in MetS patients. As opposed to proinflammatory adipokines, adiponectin levels are instead reduced in patients with excess visceral adiposity [Citation9].

Arbitrary cut-offs based on epidemiological studies have been adopted for an elevated waist circumference (WC) [Citation10] rated by all as an appropriate maker of central obesity with a strong association with the metabolic components of cardiovascular disease. However, cut-offs for WC varied for sexes, ethnic groups and even countries [Citation1,Citation3]. The IDF proposed a WC ≥94 cm for men and ≥80 cm for women as a threshold for abdominal obesity [Citation3]; these thresholds were, however, not found to be applicable to most Western populations. Higher thresholds are generally used to define abdominal obesity in the United States [Citation11] and the AHA/NHLBI (ATPIII) guidelines for MetS recognize an increased cardiovascular and diabetic risk at WCs of ≥102 cm for men and ≥88 cm for women [Citation4].

In view of the difficulty in agreeing on a common threshold, in addition to the general unavailability of an accurate WC measurement, other variables, such as BMI [Citation12,Citation13] and the waist-to-height ratio (WHtR) have been suggested. This latter, in addition to WC, is being adopted as a more accurate indicator of obesity-related cardiovascular risk. The WHtR has the advantage of being similar in most populations, i.e. generally below 0.5 for both men and women, as well as for ethnical subgroups [Citation14].

Atherogenic dyslipidemia

Carriers of MetS are characterized by a triad of metabolic perturbations called “atherogenic dyslipidemia”. Such perturbations, frequently linked to insulin-resistance (IR), the hallmark of pre-diabetes and T2D, are comprehensive of (i) high levels of apolipoprotein B (apoB)-containing lipoproteins, including very-low-density lipoprotein (VLDL) remnants, (ii) small low-density lipoprotein (sLDL), and (iii) reduced high-density lipoprotein (HDL) levels [Citation15]. In particular, IR affects the activities of enzymes and transfers proteins that play a key role in lipoprotein metabolism, such as lipoprotein lipase (LPL) and cholesteryl ester transfer protein (CETP). IR may also affect hepatic lipase (HL), phospholipid transfer protein (PLTP), and endothelial lipase (EL) [Citation16].

IR may cause these metabolic changes in the presence of suppressed hormone-sensitive lipase (HSL) in visceral adipocytes; this process raises free fatty acids (FFA) mobilization from adipose tissue, leading to hepatic triglyceride (TG) overload [Citation17]. HSL is an intracellular neutral lipase that hydrolyzes triacylglycerols, diacylglycerols, monoacylglycerols and cholesteryl esters. The increased efflux of FFA from adipose tissue and their reduced skeletal muscle uptake raises FA flux to the liver [Citation18], thus raising VLDL secretion and consequent plasma TG levels [Citation19].

Insulin may further affect TG-rich lipoprotein metabolism by inhibiting the microsomal transfer protein (MTP), responsible for the lipidation of nascent apoB during their translocation into the lumen of the endoplasmic reticulum [Citation20]. Notably, TGs lipidating apoB are derived from the FFA released from the adipocyte, from VLDL remnants and from de novo lipogenesis [Citation21]. The reduced activity of LPL [Citation22], impairing the metabolism of TG-rich lipoproteins, may lead to hypertriglyceridemia and will, in turn, increase the CETP-mediated exchange of VLDL TG for HDL-CE. This step will raise the atherogenic cholesterol-rich VLDL remnant particles and the TG-rich, cholesterol-depleted HDL particles [Citation23].

Specifically, in the condition of “atherogenic dyslipidemia” excess TGs in HDL become a substrate for HL and LPL, enzymes that convert HDL to smaller particles filtered by the renal glomeruli and degraded in renal tubular cells [Citation24]. Besides the decrement in HDL concentrations, in the presence of hypertriglyceridemia, the higher activity of cholesteryl ester transfer protein (CETP) accounts for a marked reduction of cholesterol carried in LDL [Citation25]. sLDL particles are rated as being more atherogenic, easily undergoing glycation and oxidation, compared with the larger LDL. sLDL (i) possess a higher capacity to penetrate the arterial wall, (ii) show prolonged plasma half-life, and (iii) lower affinity for the LDLR [Citation26].

Thus, a point to note is that in carriers of MetS, LDL-C is not the only lipid goal: the 2016 European guidelines point out to monitor atherogenic TG-rich lipoproteins (VLDL, IDL and remnants), recommending the achievement of specific cut-offs for non-HDL-C and apoB [Citation27]. Indeed, non-HDL-C may be used to estimate the total amount of atherogenic lipoproteins in plasma, being well related to apoB levels.

Elevated blood pressure

High blood pressure is one of the most relevant independent risk factors for cardiovascular disease; indeed, worldwide, suboptimal blood pressure accounts for around 7.6 million premature deaths annually [Citation28]. At ages 40–69 years, a reduction of 20 mmHg in systolic blood pressure (SBP), halves the risk of coronary heart diseases [Citation29]. Based on the European guidelines, hypertension is defined as values >140mmHg SBP and/or >90mmHg DBP, whereas a SBP goal <140 mmHg is recommended in patients with diabetes. Moreover, a diastolic blood pressure (DBP) target of <90 mmHg is always recommended, except in patients with diabetes, in whom the recommended objective is reduced to <85 mmHg [Citation30]. A recent meta-analysis of randomized trials of blood pressure-lowering agents in >100,000 patients with T2D confirmed that lowering blood pressure reduces the risk of cardiovascular events, coronary artery disease, stroke as well as of all-cause mortality [Citation31].

Most recently there has been a heated debate on the blood pressure target values. The SPRINT study on drug treated hypertensives indicated that an SBP below 120 mmHg should be the appropriate target for an optimal risk reduction [Citation32]. This was not accepted by other investigators [Citation33], but a most recent large-scale study indicated that outcomes after therapy may differ according to the achieved blood pressure targets in specific patient subgroups [Citation34] and the lowest targets may not always be the most appropriate. In MetS, blood pressure elevation is linked to IR, possibly to the elevated WC. It is a common practice to note that blood pressure reduction occurs in the presence of reduced body weight and improved glucose control [Citation35].

Elevated glycaemia

Elevated plasma glucose as well as increased immunoreactive insulin are in general the major features of what was earlier called “chemical diabetes” [Citation36]. Increased insulin response to an elevated glucose has been, from then on, generally linked to a higher cardiovascular risk, in particular hyperinsulinemia being found in the majority of hypertensives and constituting a common feature of the triad: obesity/glucose-intolerance/hypertension [Citation37]. Hyperglycemia per se leads to an increased risk of reduced fibrinolysis and T2D [Citation38]; eventually leading to a definition of elevated fasting glucose as a risk factor in the Framingham OFFSPRING Study [Citation39].

In view of the poor practicality of evaluating IR or even insulin levels per se, investigators aimed to define an optimal correlation between glycaemia and IR to obtain a reliable indicator of the presence of MetS. Initially, high significance was given to the condition of “impaired fasting glucose” (IFG), i.e. a fasting glycaemia above 110 mg/dl [Citation40] and the best threshold for fasting glucose based on correlations between glucose and insulin was 110–115 mg/dl [Citation41]. Later, it was suggested that this fell short of properly diagnosing a syndrome characterized by hyperinsulinaemia, and an adequate information could be provided by a fasting glycaemia above 100 mg/dl, resulting in the classification of 20.4% of patients in their study group as IR, with a predictive value of 63% [Citation42].

A fasting glucose ≥100 mg/dl has thus been selected as the basis for the diagnosis of MetS. The possible overlap between IFG and MetS has been clearly outlined: in a non-diabetic population ≥50 years of age, about twice as many individuals have IFG and MetS versus those with IFG alone Grundy [Citation43]. A sizable portion of non-diabetics have MetS without IFG and vice versa. With these considerations, it appears that the MetS is a pre-diabetic state and MetS treatment of hyperglycaemia should aim to the prevention of microvascular disease [Citation44]. In the absence of specific treatments aimed only to glucose reduction, priority must be given to reduce IR. The efficacy of this approach may be lifestyle intervention, weight reduction and increased physical activity as well established by the results of the DPP trial [Citation45].

Nutraceutical approach to metabolic syndrome

Functional foods: plant proteins

Functional foods are characterized by the presence of components with a clear activity on metabolite levels that may be associated to specific clinical syndromes [Citation46]. In the case of MetS, plant proteins deserve a special consideration. Indeed, protein-enriched diets have become a popular strategy to enhance weight management and weight loss [Citation47]. Increasing protein intake may also have a potential for the prevention of T2D [Citation48]. Although optimal amount and quality of proteins for these indications are still controversial, there are strong indications that, e.g. high intake of red meat is associated with an increased risk of T2D [Citation49], whereas plant proteins are generally associated with some improvement in diabetes risk and also in the general management of hyperlipidaemia.

Very recently, Virtanen et al. [Citation50] in a follow up of the Kuopio Ischemic Heart Disease Risk Factor Study clearly indicated that plant proteins are associated with a decreased risk of T2D. Data on MetS are less extensive but two types of proteins, i.e. lupin and soy, have been studied more in details in patients with MetS providing clinical evidence of potential benefit [Citation51]. In addition, other plant proteins, in particular pea and wheat protein, have shown some potential benefit.

Lupin

Lupin is a protein-rich grain legume. It commonly represents four domestic species, i.e. Lupinus albus (white lupin), L. luteus (yellow lupin), L. mutabilis (pearl lupin) and L. angustifolius (sweet leaf lupin). In Europe, both L. angustifolius, available as a protein concentrate, and L. albus are widely available. Lupin seeds have the interesting feature of a limited, close to nil, content of antinutritional factors such as phytate, tannins, lectins, protease inhibitors and oligosaccarides [Citation52]. Lupin seeds contain two classes of proteins, i.e. the 7S and 11S globulins, (also conglutins α and β) in addition to two minor distinct proteins, conglutin γ and δ..

Lupin proteins have been studied for a number of years mainly for their activity on plasma cholesterol reduction, attributable in large part to an LDL-receptor (LDL-R) activating mechanism [Citation53]. Moreover, conglutin γ has been shown to reduce glycemia both in animal models and, moderately, in humans, particularly in the postprandial condition [Citation54]. In animal models, similar to soy proteins, lupin proteins have displayed hypolipidemic and a remarkable antiatherosclerotic effect [Citation55].

In order to investigate the mechanism/s of the possible activity on plasma glucose, recently different soy and lupin proteins were hydrolyzed by pepsin and trypsin and the resulting peptides separated and screened for their capacity to inhibit dipeptidyl peptidase IV (DPP-IV). This new molecular target is correlated to the development of T2D; a number of pharmacological inhibitors have been developed and are currently being used [Citation56]. In the post-hydrolysis study [Citation57], two small peptides were isolated and identified as Soy 1 and Lup 1, both with efficient inhibitory capacity on DPP-IV, with IC50s equal to 106 and 228 μm, respectively, thus indicating that both proteins may be sources of DPP-IV inhibitory peptides.

Clinically, lupin proteins have been tested in different conditions, predominantly in hypercholesterolemia. Within this context, possibly due to the mechanism exerted on the LDL-R, a positive effect on LDL-C and on the LDL:HDL cholesterol ratio was reported in two studies, i.e. one with supplementation [Citation58] and another with a mixed diet enriched with lupin [Citation59]. Our group showed instead that lupin protein combinations with cellulose led to a remarkable hypocholesterolemic effect; a similar range of cholesterolaemia reduction was noted after a pea protein/fibre combination [Citation60].

A TG reduction was noted in rodent studies, apparently associated with reduced fatty acid synthesis and increased TG hydrolysis [Citation61]. This was not noted in studies in hypercholesterolemic individuals. However, in a randomized study in patients predominantly with MetS, Pavanello et al. [Citation62] reported that a lupin protein concentrate, compared with a lactose-free milk powder diet, led to a reduction, in addition to the LDL cholesterol (−8.0%), also of non-HDL cholesterol (−7.5%). This study also reported that lupin proteins, in men, lead to a 12.7% reduction of proprotein convertase subtilisin/kexin type 9 (PCSK9), the key regulator of LDLR [Citation63,Citation64]. A direct modulation of the protein–protein interaction of PCSK9 with the LDL-R has been shown [Citation65].

Further, lupin proteins can provide a potential T2D treatment, possibly also due to the recently described modulatory effect of conglutyn β on the insulin signaling pathways [Citation66]. It can be, thus, concluded that lupin protein supplemented diet or a well-designed whole diet with lupin proteins can provide an effective dietary tool for the management of the biochemical alterations characteristic of MetS.

Soy proteins

Soy proteins are the most widely evaluated dietary proteins for metabolic control. Proteins from Glycine max are the prototype plant proteins and, as such, have reached the attention, as reported in the recent systematic review on the effects of plant versus animal protein sources on features of MetS [Citation51].

In addition to proteins, glycine max provides components such as isoflavones or phytoestrogens, with a potential estrogenic activity and possibly antiatherosclerotic effects [Citation67]. Isoflavones may be of potential risk for, particularly growing children, but, so far, there is no clear evidence of a significant activity on the major components of MetS [Citation68]. Indeed, careful extraction of isoflavones from soy did not change their lipid lowering and LDL-R stimulating activity in an experimental animal model [Citation69]. While there is clear evidence that soy proteins per se can raise the activity of LDL-R in different animal models and in familial hypercholesterolemic patients [Citation70], decreased triglyceridaemia and changes in other MetS-associated variables have been reported in a lower number of studies.

Some reports have shown an improvement of glucose homeostasis after soy proteins. In post-menopausal women with MetS, soy proteins and soy nuts were compared (nuts have somewhat reduced protein and slightly higher phytoestrogen content). The soy nut regimen reduced fasting glucose more significantly than the soy protein, in addition to an improved LDL-cholesterol reduction (−5.0 ± 0.6% versus protein and −9.5%±0.6% versus control diet, both p <.01) [Citation71]. The HOMA index was also reduced, somewhat more, by soy nuts (−7.4 ± 0.8% versus protein and −12.9 ± 0.9% versus control diet, both p < .01).

Changes of triglyceridaemia were reported in a number of studies. In individuals with mildly elevated plasma TG, the reduction was in the range of 15–20%, independent of isoflavone content [Citation72]. In a detailed study in patients with moderate hypercholesterolemia treated with the bile acid binding resin colesevelam, 25 g/d of an insoluble fraction of partially hydrolyzed soy proteins were compared with a similar intake of milk proteins [Citation73]. There was a clear benefit of soy proteins on LDL-cholesterol (10.9 ± 1.8 versus −5.9 ± 2% with milk proteins, p < .01) and a somewhat higher rise of HDL-cholesterol. The difference in TG levels was modest, but non-HDL-cholesterol, was reduced to a highly significant extent with soy proteins (−10.8 ± 1.7% versus −3.9 ± 1.8% with milk protein, p < .01). There was also a highly significant difference in apo B levels, whereas faecal sterol excretion was reduced by milk proteins and significantly raised by soy proteins.

In a single study, a direct comparison of the acute intake of soy versus lupin protein on glycaemia was evaluated [Citation74] in T2D patients. A soy or lupin-based beverage containing 50 g glucose significantly reduced glycaemia for 4 h post- beverage, with no significant difference between lupin and soy. The insulin response was somewhat higher for lupin and soy, compared with controls.

While blood pressure changes following plant protein sources, compared with animal proteins, have been mainly inconsistent, body changes have been generally characterized by some weight reduction [Citation51]. Animal protein intake was positively associated with BMI and WC but plant proteins were instead inversely associated with BMI only in males and with WC in both genders [Citation75]. In the recent longitudinal EPIC-PANACE study [Citation76], increasing dietary plant source proteins by 5% at the expense of animal-source proteins in an isoenergetic diet reduced weight gain by nearly 1 kg per year in men over a 5-year period, not in women. In another study, in children, an increase in weight and BMI was noted after consumption of animal proteins, especially dairy (for periods of up to 7 years) versus vegetable proteins [Citation77].

A study by our group specifically investigated patients with clinical features of MetS; they received 30 g/d soy proteins in parallel group versus animal food. A significant reduction of body weight (−1.5% and BMI −1.5%) was observed [Citation78]. The expected reductions of total and LDL-cholesterol as well as of non-HDL-C (−7.14%) versus milk proteins were found (). Thirteen out of the 26 participating subjects had a reduction of MetS indexes, bringing them to normality.

Figure 1. Percentage median changes of non-HDL-C. Subjects randomly assigned to receive the experimental diet, containing whole soy foods corresponding to 30 g/day soy protein in substitution of animal foods containing the same amount of protein or the control diet containing the animal foods, for 12 weeks [with permission of Elsevier [Citation78]].

Figure 1. Percentage median changes of non-HDL-C. Subjects randomly assigned to receive the experimental diet, containing whole soy foods corresponding to 30 g/day soy protein in substitution of animal foods containing the same amount of protein or the control diet containing the animal foods, for 12 weeks [with permission of Elsevier [Citation78]].

Other plant protein sources (without isoflavones) have been tested in individuals with MetS. Among these, pea, fava bean proteins and wheat gluten did not show any further beneficial effect on fasting lipemia compared with proteins of animal sources. However, wheat gluten ingestion resulted in a larger postprandial TG response compared with wheat proteins [Citation79]. In general, studies comparing diets from plant versus animal sources with a positive metabolic effect had a longer duration [Citation51].

Probiotics, prebiotics and synbiotics

In humans, microbial density comprises a biomass of 1.5–2.0 kg and increases from the proximal to the distal end of the intestine; in the body, the ratio of bacteria to human cells is close to 1:1 [Citation80]. Recently, numerous studies in humans and in animal models have reported how gut microbiota, established within the first 3 years of life, converts dietary and endogenous molecules into metabolites, that allow communication with peripheral organs and tissues in the host [Citation81].

Among microbial metabolites, short-chain fatty acids, i.e. acetate, butyrate and propionate, are known to exert several health benefits, including reduction of the luminal pH, inhibition of enteropathogens, increased absorption of some nutrients and maintenance of gut barrier function [Citation82]. Conversely, a rise in the absorption of branched-chain amino acids, as in the case of alterations in the intestinal barrier (e.g. high-fat diets), is associated with a higher risk of T2D [Citation83,Citation84]. Indeed, dysbiosis, i.e. the alteration of gut homeostasis, is associated to obesity, T2D and IR, all traits of MetS that can lead to atherosclerosis and heart failure [Citation85]. It has been, further, proposed an “Interaction Model”, demonstrating that, at least in permissive genetic backgrounds, gut microbiota plays a significant role in modulating key metabolic traits, such as obesity, diabetes and IR [Citation86] ().

Figure 2. The development of obesity, insulin resistance, type 2 diabetes and metabolic syndrome in general are the consequence of a complex multidirectional interaction between host genetics, environment, diet and the gut microbiota [with permission of Elsevier [Citation86]].

Figure 2. The development of obesity, insulin resistance, type 2 diabetes and metabolic syndrome in general are the consequence of a complex multidirectional interaction between host genetics, environment, diet and the gut microbiota [with permission of Elsevier [Citation86]].

Since diet shapes the composition of gut microbiota, restoring its diversity and activity, nutrients and particularly nutraceuticals can represent a promising approach to improve metabolic features of MetS [Citation81]. Moreover, in older people, in which microbiota is susceptible to a larger inter-individual variation, dietary supplements with defined food ingredients promoting specific components of microbiota may be useful in maintaining the best health status [Citation87].

Consumption of some nutraceuticals i.e. probiotics, prebiotics and synbiotics should be of primary consideration. Probiotics are living micro-organisms that when administered in adequate amounts confers beneficial health effects to the host (i.e. Lactobacillus rhamnosus GG, L. reuteri, Bifidobacteria certain strains of L. casei or the L. acidophilus group) [Citation88].

Prebiotics are defined as selectively non-digestible plant-derived carbohydrates, allowing specific changes, both in the composition and in the activity of gastrointestinal microflora; indeed, these food ingredients (i.e., inulin and its hydrolysis product oligofructose, and (trans)galactooligosaccharides) confer health benefits by stimulating growth and/or activity of a limited number of bacteria within the colon [Citation89]. The definition of “prebiotic” should include (a) resistance to gastric acidity, hydrolysis by mammalian enzymes and gastrointestinal absorption; (b) fermentation by intestinal microflora; (c) selective stimulation of the growth and/or activity of intestinal bacteria associated with health and wellbeing [Citation89]. Synbiotics are a synergistic combination of pro- and prebiotics [Citation90].

A detailed summary of data from randomized controlled studies (RCTs) related to the use of these nutraceuticals should also consider that results may be affected by different biases, since the majority of the RCTs (i) did not follow the same design, (ii) were small in terms of participants, (ii) were of short duration and (iii) used probiotics from multiple strains as well as different delivery methods (i.e. yogurt, cheese or capsules) [Citation91]. Thus, RCTs studies with a variety of sources, daily doses and different durations of intervention are required to confirm health benefits and specific roles of probiotics, prebiotics and synbiotics on the metabolic components of the MetS.

Probiotics and traits of metabolic syndrome

Weight and BMI

Data from a meta-analysis of 15 RCTs reported, in obese, that the administration of 2.7*1010 Colony Forming Units (CFU)/day of Lactobacilli probiotics for 2–3 months reduced body weight (kg) by −0.54 (95% CI −0.83, −0.25; p < .001); when BMI (kg/m2) was instead considered, reduction in the Lactobacilli arms was −0.43 (95% CI −0.67, −0.20; p = .005). Conversely, upon probiotic supplementation, a slight increment in body weight was observed in children (+0.20 kg; 95% CI 0.04, 0.36) and infants (+0.30 kg; 95% CI −0.01, 0.62; p = 0.05) [Citation92]. Another meta-analysis, on four RCTs, showed no significant effects of probiotics on body weight, BMI and visceral fat of obese subjects [Citation93].

In line with the above-described findings, reporting a weak (<3%) or absent effect of probiotics on body components [Citation94], in obese with elevated cardiovascular risk, probiotic supplementation reduced BMI by 0.52 kg/m2 (95% CI −0.81, −0.25; p < .001) and waist circumference by −2.11 cm (95% CI −3.54, −0.68; p = .004) [Citation95]. A further contribution to the understanding of this complex issue has come from a meta-regression analysis reporting that the effects of probiotics on BMI depend on the duration of intervention (≥ 8 weeks), number of species of probiotics and baseline BMI ≥25 kg/m2, thus highlighting the effectiveness of probiotics in reducing BMI, especially in overweight or obese subjects [Citation96].

A possible mechanism explaining the effects on weight loss are the improvement of the intestinal barrier function that may reduce metabolic endotoxemia [Citation97], thus ameliorating levels of lipopolysaccharides (LPS). In the adipose tissue, endotoxins from LPS trigger systemic and local inflammation with an increase in reactive oxygen species (ROS) [Citation97]. As a possible associated event, bacterial fragments can increase the number of preadipocytes [Citation98]. Among other proposed mechanisms linking probiotics and body weight reduction there are the ability (i) of the microbioma to extract energy from the diet [Citation99] and (ii) to increase the secretion of the gut hormone glucagon-like peptide (GLP-1) enhancing satiety and reducing energy intake [Citation100].

Blood pressure

A meta-analysis of 9 RTCs relative to the administration of probiotics from different strains (L. reuteri, L. plantarum, L. helveticus, Enterococcus faecium and Streptococcus thermophilus), with a total daily consumption between 109 and 1012 CFU, with a duration of 3–9 weeks, showed an overall improvement in blood pressure. A reduction of −3.58 mmHg (95% CI −6.46, −0.66; p < .00001) and −2.38 mmHg (95% CI −3.84, −0.93; p < .0001) was recorded for SPB and DBP, respectively. Interestingly, both SBP and DBP fell further when a sub-analysis was conducted taking into consideration: (i) duration of treatment ≥8 weeks, (ii) BMI ≥30 kg/m2, (iii) dairy source of probiotics (versus capsules), (iv) consumption of multiple probiotic species, (v) daily dose consumption ≥1011CFU and (vi) blood pressure at baseline ≥135/85 mmHg [Citation101].

The anti-hypertensive effects of probiotic supplementation were also confirmed in subjects with T2D; reductions of 3.28 mmHg and of 2.131 mmHg in SBP and DBP, respectively, have been reported. These effects were inversely related to BMI (> 29 kg/m2) and age [Citation102].

The antihypertensive activity of probiotics has been also attributed to the reduction of inflammation-mediated endothelial dysfunction, nitric oxide synthase downregulation [Citation103], and release of angiotensin-converting enzyme inhibitory peptides. Among these, the tripeptides Val-Pro-Pro and Ile-Pro-Pro have been the most extensively investigated [Citation104]. As previously reviewed, two major commercial products, based on sour milk, i.e. the Finnish Evolus® and the Japanese Calpis® are presently available [Citation105]. However, due to the skepticism regarding their cost and possible effectiveness, they have not reached a wide distribution outside these countries.

Fasting plasma glucose

The effect of probiotic supplementation on glucose homeostasis is influenced by the basal levels of FPG (≥ 126 mg/dl), length of intervention (≥ 8 weeks), baseline BMI values (≥ 30 kg/m2), administration of multiple species of probiotics, daily dose consumption ≥1010 CFU and the selected sources, i.e. supplements and foods instead of fermented milk or yogurt. Indeed, the presence of lactose in fermented milk may raise FPG. Conversely, if the overall population is considered, a non-statistical FBG reduction of 0.17 mmol/l is observed [Citation106].

In studies conducted in cohorts of T2D patients, mean reductions of 0.61 mmol/l [Citation107] or 0.98 mmol/l were reported [Citation108]. Interestingly, the higher the number of bacterial species is, the more significant the FPG reduction is [Citation107,Citation109]. Contrasting and inconclusive data have been instead reported on the effectiveness of probiotic supplementation on HOMA-IR and glycosylated hemoglobin (HbA1C) levels, two hallmarks of IR [Citation107–109]. The reduction of low-grade inflammation promoted by gut-derived LPS and metabolic endotoxemia has been proposed as mechanisms linking probiotics and glucose metabolism [Citation110]. Indeed, serum endotoxin levels are correlated with fasting insulin [Citation111] and are two-fold higher in T2D patients compared with non-diabetics [Citation112].

HDL and TG

A recent meta-analysis of 30 RCTs, investigating the effect of different strains of probiotics (L. acidophilus, L. plantarum and L. helveticus) on lipid parameters, reported no changes in HDL-C (−0.24 mg/dl; 95% CI −1.6, 1.1) and TG (−5.3 mg/dl; 95%CI −11.1, 0.4) levels [Citation91], between subjects taking probiotics and controls. This has been confirmed in mild-to-moderate hypercholesterolemic individuals in whom probiotic supplementation was ineffective in improving HDL-C (0.01 mg/dl; 95%Cl −0.02, 0.03) and TG (0.01 mg/dl; 95%Cl −0.08, 0.09) levels; these findings were not modified upon correction for baseline BMI (≥25 kg/m2), duration (≥4 weeks), age (≥ 45 years), preparation (fermented milk products versus probiotic preparations) and bacterial strains [Citation113].

Considering that probiotics exert a hypocholesterolemic effect by reducing the intestinal absorption of cholesterol or by enhancing the bile salt hydrolase activity, a lack of efficacy in modulating HDL-C and TG, both linked to the atherogenic dyslipidemia, is expectable [Citation114]. Notably, if only T2D patients are considered, probiotic consumption reduced TG levels by -24.48 mg/dl (95%Cl −33.7, 11.2) with no HDL-C changes [Citation102]. HDL-C may be raised by 16 mg/dl if a longer duration of probiotic administration (>8 versus ≤8 weeks) is considered [Citation107].

Prebiotics, synbiotics and the major traits of metabolic syndrome

The effects of prebiotics on the metabolic traits of MetS have been evaluated in a limited number of studies investigating comparable outcomes. Their small sample sizes, heterogeneity among trial subjects, disease conditions, prebiotic supplements, intervention duration and outcomes need thus to be carefully considered.

Among the different outcomes evaluated in trials investigating the effects of prebiotic supplementation on metabolic traits, i.e. body weight, glucose homeostasis and TG, evidence indicates that dietary supplementation impacts only on post-prandial glucose (−0.76 mmol/l (95%Cl −1.41, −0.12)) and insulin (−0.77 UI/l (95%Cl −1.50, −0.04)) levels leaving the other parameters unaffected. Interestingly, although self-reported, an improvement in satiety was found upon prebiotic intake [Citation115]. Not surprisingly, prebiotics have been related to a decrement in Ghrelin levels and to an increment in peptide YY and GLP-1 secretion, all involved in appetite regulation [Citation116,Citation117].

In subjects with a BMI ≥25 kg/m2 and carriers of diabetes, evaluation of the effects of prebiotics compared with placebo showed an improvement in TG and HDL-C levels, −0.72 mmol/l and +0.49 mmol/l, respectively [Citation118]. The same authors reported that a synbiotic supplementation decreases plasma fasting insulin and TG levels by −0.39 UI/l (95%Cl −0.75, −0.02) and −0.43 mmol/l (95%Cl −0.70, −0.15), respectively. No changes in FPG were noted [Citation118], similar to what reported in a very recent meta-analysis [Citation106].

Vitamin D

Vitamin D has generally been shown to exert a minor role in the control of MetS. However, recent data have indicated vitamin D levels as an important variable, underlying, in some cases, MetS, in addition to being a possibly significant determinant of cardiovascular risk.

Vitamin D has a well-known primary physiological role in regulating calcium homeostasis. However, besides bone metabolism, growing evidence has described the involvement of vitamin D on glucose metabolism, blood pressure and other metabolic traits related to MetS [Citation119].

Being mainly synthesized in the skin upon ultraviolet-B radiation, vitamin D is metabolized by the hepatic 25-hydroxylase enzyme to 25-hydroxyvitamin D [25(OH)D] and by a renal 1α-hydroxylase to the vitamin D hormone calcitriol, also called 1,25-(OH)2 vitamin D; the active form of vitamin D [Citation120]. Since 1,25-(OH)2 vitamin D is active for a maximum of 27 h, 25(OH)D levels are the landmarks for the assessment of vitamin D status [Citation121,Citation122]. Specifically, when circulating levels of 25(OH)D are ≤30 nmol/l, this is diagnosed as a condition of vitamin D deficiency; inadequacy is when 25(OH)D levels are between 30 and 50 nmol/l and sufficiency when concentrations are in the range of 50–75 nmol/l [Citation123].

The relationship between 25(OH)D levels and risk of MetS has been recently reviewed; a dose–response meta-analysis of 16 cross-sectional studies showed a linear and inverse association between the two variables. Moreover, the risk-ratios of MetS, per 25 nmol/l, increment in the serum/plasma 25(OH)D levels, were 0.87 (95%Cl 0.83–0.92). Albeit, somewhat attenuated, this effect was maintained upon correction for sex, age (middle versus elderly), latitude (high versus low), assay detection method, criteria of MetS definition [Citation124]. The same results were obtained when the relative risk for MetS was assessed comparing individuals in the top versus bottom tertiles of baseline 25(OH)D levels [Citation119]. In the context of MetS, obese individuals show a decrement in circulating 25(OH)D, i.e. of 0.27 ng/ml in vitamin D3 for each 1 kg/m2 increment in BMI [Citation125]. Among the different suggested hypotheses [Citation126], the concept of reverse causality is to be taken into consideration. Indeed, if an inverse correlation between obesity and vitamin D levels is true, on the other hand, vitamin D may be sequestered or stored in the adipose tissue leading to lower 25(OH)D serum levels. Thus, obesity may be a cause and not a consequence of vitamin D deficiency [Citation127].

An inverse association between baseline 25(OH)D levels and incidence of T2D has been also reported. Comparing the highest to the lowest cutoffs of 25(OH)D levels, a relative risk of 0.62 (95%Cl 0.54–0.70) emerged from a pooled analysis of 21 prospective studies. This finding was not affected by sex, duration of follow-up, sample size, diabetes or 25(OH)D assay method [Citation128]. The same conclusions were reached by Khan et al. showing how individuals in the top versus bottom tertile of baseline 25(OH)D blood levels had a relative risk of 0.81 (95%Cl 0.71–0.92) for T2D incidence [Citation119]. Conversely, when vitamin D supplementation has been taken into consideration, data from RCTs did not show any effect on fasting glucose, HbA1C, insulin resistance or incidence of diabetes [Citation129]. In general, vitamin D may have a direct effect on beta-cell function through the activation of the vitamin D receptor expressed in beta-cells; lack of a functional vitamin D receptor leads to an impairment in glucose metabolism, although a vitamin D-mediated beta-cell survival should not be excluded [Citation129].

The earliest evidence linking vitamin D to blood pressure came from the observation that low UVB exposure was associated with an increased hypertension risk [Citation130]. Among the 12,644 participants of a cross-sectional survey, mean SBP and DBP were 3.0 mmHg and 1.6 mmHg lower for people in the highest 25(OH)D quintile (≥ 85.7 nmol/l) compared with the lowest (<40.4 nmol/l) [Citation131]. This finding has been confirmed by showing an improvement in blood pressure directly related to the increment in serum 25(OH)D levels; upon correction for sex, age, BMI and physical activity, the differences in SBP and DBP between the lowest (<41.4 nmol/l) and highest (>62.6 nmol/l) serum 25(OH)D quartiles were 3.6 and 1.0 mmHg, respectively [Citation132].

A meta-analysis on 283,537 subjects, examining the association between vitamin D status and hypertension risk, showed that individuals in the top 25(OH)D tertile have a 30% lower risk of developing hypertension compared with those in the lowest tertile [0.70 (95%Cl 0.58–0.86)]. Notably, a 10 ng/ml increment in baseline vitamin D levels corresponded to a 12% reduction in risk [0.88 (95%Cl 0.81, 0.97)] [Citation133]. In this type of correlations, the importance of baseline 25(OH)D levels are confirmed also for very low reference values; subjects with circulating levels <15 ng/ml compared with those with levels ≥30 ng/ml have a 3.18 (95%Cl 1.39–7.29) higher relative risk of hypertension development [Citation134].

Overall, the antihypertensive effects of vitamin D may depend on a direct activation of the vitamin D receptor on vascular endothelial and smooth muscle cells or on a modulation of the renin–angiotensin–aldosterone system [Citation135,Citation136].

Conversely, different conclusions were reached when the vitamin D supplementation was used as a blood pressure-lowering agent. Indeed, no evidence of blood pressure reduction upon vitamin D supplementation was seen on either SBP (0.0 mmHg (95%Cl –0.8 to –0.8)) or DBP (0.1 mmHg (95%Cl –0.6 to −0.5) [Citation137]. Similar results were obtained after a sub-analysis for baseline SBP >140 mmHg or ≤140 mmHg and for baseline 25(OH)D values >20 ng/ml or ≤20 ng/ml. Supplementation with vitamin D (50,000 IU/week) in MetS patients for 16 weeks significantly reduced levels of the vascular adhesion molecule-1 and of E selectin. However, there was no significant activity on high-sensitivity C-reactive protein and carotid intima media thickness [Citation138].

Curcumin

Curcuminoids are polyphenolic compounds derived from the dried rhizomes of Curcuma longa L. (turmeric) and are responsible for the orange-yellow colour [Citation139]. Curcumin is the major component, i.e. constituting about 5% of turmeric, with the structural analogues demethoxycurcumin and bis demethoxycurcumin. Unstable at physiological pH, it rapidly degrades to oxidized products, which mediate topoisomerase poisoning as well as other antioxidant, anti-inflammatory and lipid lowering effects [Citation140].

Among the multiple pharmacodynamic effects of curcumin are the well-established antioxidant and anti-inflammatory properties, demonstrated by in vitro and in vivo studies. Indeed, this compound proved to increase serum total antioxidant capacity and superoxide dismutase, together with an increase in glutathione concentrations and reduction in lipid peroxides [Citation141,Citation142]. Interestingly, curcumin can also regulate cytokines, protein kinase, adhesion molecules and other enzymes (TNF-α, IL-1, IL-6, TGF-β, MCP-1, etc.) [Citation139,Citation143,Citation144]. Supplementation with curcuminoids was associated with a statistically significant reduction in circulating high-sensitivity (hs)-CRP levels in a meta-analysis (weighed mean difference: −6.44 mg/l; 95% CI: −10.77 to −2.11) [Citation145]. A small trial on 14 males showed a significant increase in postprandial flow-mediated dilatation (FMD) (from 5.2% to 6.6%) after a single consumption of curry meal containing curcumin [Citation146], suggesting a potential role in preventing endothelial dysfunction.

Among the multiple properties on a wide range of diseases, curcumin proved to modify almost all features of MetS [Citation147]. The main metabolic effect of curcumin seems to be related to an activity as insulin sensitizer [Citation148,Citation149], well demonstrated in animal models of diabetes [Citation148], and confirmed in human studies. In a double-blind randomized trial, pre-diabetic patients achieved an improvement of all markers of insulin sensitivity (C-peptide, HOMA-IR and HOMA for β cells) after 9 months with 1500 mg of curcumin compared with placebo [Citation150]. The curcumin extract in the same study also prevented T2D development (0% of curcumin-treated developed DM versus 16.4% of placebo-treated patients). A +22.5% increase in adiponectin levels was also detected, later confirmed in a further meta-analysis reporting a 77% rise (95% CI: 6.14–147.42) together with a 26% decrease in leptin (95% CI: −70.44-17) [Citation142]; both adiponectin and leptin have been linked to cardiovascular diseases [Citation7]. A shorter treatment period provided no comparable benefit on glucose homeostasis [Citation151], with, however, a significant improvement of HDL-cholesterol (+6.8%) and a reduction of TG (−28.8) and of non-HDL-C (−13.6%). A larger study on 117 subjects with MetS provided confirmatory findings, indicating, in addition a significant antioxidant and anti-inflammatory activity with a marked reduction (−2.2 mg/l) of CRP concentrations [Citation152].

The lipid-lowering mechanisms of curcuminoids seem to reside in their ability to increase cholesterol efflux via ABCA1 and APOA-I expression [Citation153,Citation154] and to inhibit the expression of Niemann–Pick C1-Like 1 (NPC1L1) via the Sterol regulatory element-binding protein 2 (SREBP2) transcription factor [Citation155]. Curcumin also downregulates PCSK9 mRNA levels by up to 31–48% in different cell lines, thus promoting LDL-R expression on the cell surface and LDL-C uptake [Citation156]. The mechanism of down-regulation of PCSK9 appears to be linked to the inhibition of the HNF-1α transcription factor: this may lead to an additional anti-inflammatory activity [Citation157]. Curcumin further demonstrated protection against atherosclerosis in LDLR−/− mice [Citation154], by modifying peroxisome proliferator-activated receptors (PPAR)-α and γ, CETP and LPL expression, therefore, affecting synthesis and catabolism of fatty acids [Citation154,Citation158].

Limitation to a wider use of curcumin is the poor oral bioavailability, representing a major obstacle to clinical development. Although usually well tolerated and safe, curcumin has a low hydrosolubility and undergoes a rapid gastric and intestinal metabolism. A number of new formulations have been described. A formulation with a hydrophilic carrier, cellulose base and natural antioxidants [Citation159] proved to be absorbed at least 30 times better than curcumin base. Recently, a formulation of curcumin encapsulated in milk exosomes proved to have improved resistance to human digestion and improved cell permeability [Citation160]. A formulation with piperine, an inhibitor of hepatic and intestinal glucoronidation is in advanced development, and the initial clinical tests have given a positive outcome [Citation161]. Development of a curcumin product with an improved bioavailability will probably lead to a better activity on the blood lipid profile, as shown in a study investigating in parallel lipid responses and blood levels [Citation162].

Berberine

It is a natural plant alkaloid, isolated from the Chinese herb Coptis kinensis (Huanglian). In the Far East, it is commonly used for diarrhoea but a potential glucose lowering effect was noted by Chinese medicine way back in the ‘80s. The breakthrough finding was the description of a powerful cholesterol lowering activity (berberine 500 mg bid for 3 months) in hypercholesterolemic patients [Citation163]. This activity was associated with an elevation of the LDL-R expression, independent of the sterol regulatory element binding proteins (SRBPs), but consequent to the activation of an extra signal regulated kinase (ERK). In a later study, Lee et al. [Citation164] reported that berberine, in addition to stabilizing hepatic LDL-R mRNA in an ERK-dependent way, could increase the transcriptional activity of LDL-R promoter by activating the JNK pathway (). Moreover, in endothelial cells, berberine has been reported to counteract pro-atherogenic and inflammatory stimuli induced by oxLDL and TNF-alpha, a mechanism involving the inhibition of lectin-like OxLDL receptor 1 and the modulation of AMPK and ERK1/2 pathways [Citation165].

Figure 3. Main mechanisms of action of berberine on the components of metabolic syndrome. AMPK: 5' AMP-activated protein kinase; GLP-1: gut hormone glucagon-like peptide; LDL: low-density lipoprotein; PCSK9: proprotein convertase subtilisin/kexin type 9; SREBP-2: sterol regulatory element-binding transcription factor 2 [adapted with permission of Elsevier [Citation205]].

Figure 3. Main mechanisms of action of berberine on the components of metabolic syndrome. AMPK: 5' AMP-activated protein kinase; GLP-1: gut hormone glucagon-like peptide; LDL: low-density lipoprotein; PCSK9: proprotein convertase subtilisin/kexin type 9; SREBP-2: sterol regulatory element-binding transcription factor 2 [adapted with permission of Elsevier [Citation205]].

These findings stimulated interest in the possible use of berberine in patients with the more classical LDL-R mutations, but Kong et al. findings were not widely confirmed, mainly because berberine was, from then on, mainly given in association with other nutraceuticals affecting cholesterolemia. Instead, the earlier traditional findings reporting an improvement of T2D were confirmed by a number of investigations. At a similar dose, as in the early study [Citation163], patients with newly diagnosed T2D, showed a similar reduction of glucose, hemoglobin A1c and TG; remarkably, fasting insulin and the HOMA index were reduced by 28.1 and 44.7% [Citation166].

The mechanism/s of berberine in addressing lipid targets, particularly, those strictly related to MetS have been extensively evaluated. Huang et al. [Citation167], working on mechanisms related to an improved insulin sensitivity, reported that berberine inhibited 3T3L adipocyte differentiation, through the transcription factors PPARγ and c/eBPα, also inhibiting the full length PPARγ, PPAR α and their target genes involved on glucose homeostasis and lipid metabolism [Citation168]. In this way, berberine may exert a weight reduction in addition to hypolipidaemic and hypoglycaemic activity.

A comparative study, in patients with T2D, on 13 weeks of berberine treatment (500 mg tid) versus a similar dose of metformin, reported similar hypoglycemic and HBA1c reducing effects of the two agents, in addition to a 21% TG reduction with berberine, versus no effect of metformin [Citation166]. In the follow-up, 48 adults with poorly controlled T2D, receiving a similar berberine supplementation, showed a marked decrease of HBA1c (from 8.1% to 7.3%), insulin and HOMA-index; in addition, TG, total and LDL-C were significantly reduced. In a large series of 106 patients with T2D and dyslipidaemia, berberine (1 g/day) for 3 months significantly reduced glycaemia, cholesterolemia, trygliceridemia and, in addition, improved the glucose disposal rate, as assessed by a hyperinsulinemic euglycemic clamp [Citation169].

The specific activity of berberine in patients with MetS has been complemented by two studies, one in animals and one in humans, both in conditions of IR. Lee et al. [Citation170] in two models of IR, i.e. the obese db/db mice and the high fat-fed Wistar rats, reported a clear IR improving effect, associated with marked body weight reduction. The authors also indicated a possible increase of the AMP-activated protein kinase (AMPK) in adipocytes and increased GLUT4 translocation in L6 cells. The potential mechanisms of the improved insulin sensitivity were well evaluated in MetS patients. After receiving 12 weeks of treatment with berberine 300 mg tid, these patients showed a significantly decreased BMI (from 31.5 ± 3.6 to 27.4 ± 2.4 kg/m2) and leptin levels (8.01 versus 5.12 μg/l both p< .001), as well of the leptin/adiponectin ratio and HOMA Index [Citation171]. These findings clearly indicate that insulin sensitivity is improved by berberine by adjustment of the adipokine secretion, both in primarily cultured adipocytes and in MetS patients. A potential additional mechanism in improving IR is by way of inhibited adipose tissue lipolysis. The antilipolytic activity is exerted by an increased adipose tissue phosphodiesterase (PDE), leading to reduced cAMP and inhibited activation of the HSL [Citation172].

The improvement of IR and the activation of AMPK, this latter similar to the mechanism of metformin [Citation173], albeit with some contrasting findings [Citation174], are certainly in line with a significant activity of the agent in MetS. Further findings have suggested that berberine can provide an effective association with cholesterol lowering drugs. Berberine reduces the liver expression of PCSK9, a serine protease favouring degradation of the LDL-R [Citation175], increased after statin therapy [Citation176]. The reduction of PCSK9 mRNA and protein levels [Citation177] are dependent on a down regulation of the hepatocyte nuclear factor-α (HNF-1α), one of the essential cofactors for the transcriptional regulation of PCSK9 [Citation178,Citation179] and responsible for the regulation of a variety of inflammatory conditions. Inhibited HNF1-α transcription has also been reported after curcumin, possibly leading to a similar reduction of PCSK9 [Citation156,Citation157].

Finally, berberine has a potentially significant activity in upregulating the reverse cholesterol transport, when this is inhibited by the CETP inhibitor torcetrapib. In this condition, inhibited CETP in dyslipidemic hamsters does not lead to a stimulation of reverse cholesterol transport, whereas this is markedly stimulated in the presence of berberine [Citation180].

The wide availability, relatively low cost and generally adequate tolerability of berberine, make it an important therapeutic addition to the list of nutraceuticals affecting MetS [Citation181]. Berberine has unfortunately a relatively low bioavailability, making it necessary to administer elevated doses, with consequent possible gastrointestinal side effects. Since the reduced bioavailability is consequent to berberine being P-gp substrate [Citation182], association of berberine with a the P-gp inhibitor Silybum marianum extract, apparently improved the lipid activity on lipid and on insulin secretion [Citation183].

Red yeast rice (RYR)

RYR is obtained by the fermentation of the Monascus purpureus yeast in rice (Oryza sativa) and is commonly used in oriental cuisine. The fermentation process enriches rice in bioactive components, including polyketides such as monacolins (compactin, monacolin K, M, L, J, X) with documented lipid-lowering effects as inhibitors of THE 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase [Citation184]. The primary monacolin in RYR is monacolin K, with the same chemical structure as lovastatin and, for this reason, classified as a drug by the US Food and Drug Administration (FDA) [Citation185]. The European Food Safety Authority (EFSA) maintains instead the use of monacolin K from RYR preparations as a nutraceutical that can reduce elevated LDL-C concentrations [Citation186].

Beneficial vascular effects in MetS can go beyond cholesterol reduction since 1200 mg/d of RYR showed endothelial protection in 50 coronary patients after 6 weeks of treatment, significantly reducing fasting hsCRP (−50% versus 25.4%) and improving post-prandial FMD compared with placebo (7.91 ± 3.16% versus 5.34 ± 2.78%) [Citation187]. Confirming these findings, Cicero et al. found a significant improvement in endothelial function, measured as pulse volume (PV) and arterial stiffness, measured as pulse wave velocity (PWV) in 40 moderately hypercholesterolemic subjects. After 4 weeks of monacolin K 10 mg treatment, PV rose by 6.0% (compared with 0.3% with placebo) and PWV decreased by 4.7% (1.1% with placebo) [Citation188]. These data could be possibly in line with a reduction in plasma concentrations of the matrix metalloproteinases (MMPs), MMP-2 (−28.05%) and MMP-9 (−27.19%) observed by the same authors in a previous clinical study [Citation189]. These effects could be consequent to an up-regulation of eNOS expression and inhibition of oxidative stress in vascular endothelia, as shown in rodents [Citation190].

There is limited evidence, i.e. from a small randomized clinical trial, of a blood pressure lowering effect of 10 mg monacolins (50% monacolin K), combined with 9 mg of hydroxytyrosol, in 50 MetS subjects [Citation191]. A significant drop in SBP and DBP by 10 and 7 mmHg, respectively, compared with placebo, was recorded after 8 weeks. So far, this effect has not been reproduced and a meta-analysis from 21 trials showed only a significant effect on SBP [Citation192].

RYR is currently used predominantly in combination with other nutraceuticals, allowing us to reduce daily doses, and exerting effects beyond cholesterol reduction. Among others, association of RYR with berberine proved to be highly effective in reducing TC by 0.68 mmol/l and LDL-C by 0.61 mmol/l, while raising HDL-C by 0.07 mmol/l, reducing TG and glucose by 0.16 mmol/l and 0.14 mmol/l, respectively [Citation193]. Interestingly, unlike statins [Citation194], this nutraceutical combination improved the leptin-to-adiponectin ratio (−17.8%), without changing adiponectin levels [Citation195]. Moreover, the same association led to an improved endothelial function and PWV [Citation189].

In addition, RYR (200 mg), in combination with berberine (500 mg) and policosanols (10 mg), has been shown to improve insulin sensitivity in patients with IR, reducing the HOMA-IR by 24% [Citation196].

Some studies assessed the efficacy of RYR in reducing cardiovascular risk in adults and elderlies in secondary prevention. Lu et al. [Citation197] carried out a prospective study on 5000 Chinese patients with a previous history of myocardial infarction, randomized to RYR or placebo and followed for an average of 4 years. RYR reduced the incidence of coronary events (5.7% versus 10.4% in the placebo group (p = .001); deaths from coronary heart disease increased by 30% (p = .04), and all-cause mortality by 33% (p = .01). These findings were later confirmed by a reduction (−43%) of coronary events and all-cause mortality (−35.8%) in 2704 hypertensive patients [Citation198].

In addition to the potential of induced statin-like CK elevations and myalgia [Citation199] after prolonged treatment [Citation200], attention should be given to the presence of citrinin, a nephrotoxic mycotoxin metabolite, derived from the fermentation process of Monascus. For this reason, EFSA has defined a maximum concentration to be taken per day of 0.2 µg/kg b.w. [Citation186]. Found in low quality RYR products, citrinin can pose a health risk as it may be mutagenic, as found in animal models, and genotoxic to human lymphocytes [Citation201,Citation202], and may also cause kidney failure in animals, although acute human toxicity is a rare event.

Conclusions

The management of MetS is a major target in atherosclerosis prevention. While treatment of hypercholesterolemia can be successfully handled using drugs affecting in particular cholesterol biosynthesis, in the case of MetS there are a number of diverse metabolic abnormalities more difficult to pursue. In particular, in MetS, insulin resistance appears to be a major abnormality that may be only partially handled by drug treatments. Insulin resistance may of course translate into altered body structure, elevated blood pressure and other metabolic and non-metabolic changes.

The very high number of carriers of MetS makes it mandatory to achieve treatment by more widely available nutritional approaches. Nutraceuticals can, thus, offer a significant help in handling MetS patients [Citation203]. They may be part of lifestyle recommendations and may include Mediterranean diet components, olive oil and its anti-oxidant components, natural legumes and cereals [Citation204]. However, in the daily management of patients, a dietary counselling alone may not be adequate. In this manuscript, we have reviewed highly standardized nutraceuticals that go from functional food components, in particular from plant proteins (e.g. soy and lupin) to probiotics and prebiotics, providing significant benefit by modifying the intestinal microbiome, to natural compounds, frequently achieving a drug-like status, e.g. red yeast rice, berberine and curcumin, as well as vitamin D (). All these nutraceuticals are easily accessible both to physicians or patients and can easily enter a lifestyle modification for improvement of MetS. Adequate formulations will certainly raise their already large medical use once bioavailability of some of these natural molecules is improved; eventually making them more widely acceptable for the management of common and high-risk metabolic and vascular abnormalities.

Table 2. Effects of nutraceuticals on the features of metabolic syndrome: evidence from randomized-controlled trials and meta-analysis.

Disclosure statement

The authors report no declarations of interest.

References

  • Expert Panel on Detection E, Treatment of High Blood Cholesterol in A. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA. 2001;285:2486–2497.
  • Einhorn D, Reaven GM, Cobin RH, et al. American College of Endocrinology position statement on the insulin resistance syndrome. Endocr Pract. 2003;9:237–252.
  • Alberti KG, Zimmet P, Shaw J. Group IDFETFC. The metabolic syndrome-a new worldwide definition. Lancet. 2005;366:1059–1062.
  • Alberti KG, Eckel RH, Grundy SM, et al. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation. 2009;120:1640–1645.
  • Bjorntorp P. “Portal” adipose tissue as a generator of risk factors for cardiovascular disease and diabetes. Arteriosclerosis. 1990;10:493–496.
  • Abbasi F, Brown BW Jr, Lamendola C, et al. Relationship between obesity, insulin resistance, and coronary heart disease risk. J Am Coll Cardiol. 2002;40:937–943.
  • Ruscica M, Baragetti A, Catapano AL, et al. Translating the biology of adipokines in atherosclerosis and cardiovascular diseases: gaps and open questions. Nutr Metab Cardiovasc Dis. 2017;27:379–395.
  • Weisberg SP, McCann D, Desai M, et al. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808.
  • Cote M, Mauriege P, Bergeron J, et al. Adiponectinemia in visceral obesity: impact on glucose tolerance and plasma lipoprotein and lipid levels in men. J Clin Endocrinol Metab. 2005;90:1434–1439.
  • Lean ME, Han TS, Morrison CE. Waist circumference as a measure for indicating need for weight management. BMJ. 1995;311:158–161.
  • Consultation W. Obesity: preventing and managing the global epidemic: introduction. Who Tech Rep Ser. 2000;894:1–253.
  • Consultation WHOE. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet. 2004;363:157–163.
  • Pischon T, Boeing H, Hoffmann K, et al. General and abdominal adiposity and risk of death in Europe. N Engl J Med. 2008;359:2105–2120.
  • Mombelli G, Zanaboni AM, Gaito S, et al. Waist-to-height ratio is a highly sensitive index for the metabolic syndrome in a Mediterranean population. Metab Syndr Relat Disord. 2009;7:477–484.
  • Ginsberg HN, Huang LS. The insulin resistance syndrome: impact on lipoprotein metabolism and atherothrombosis. J Cardiovasc Risk. 2000;7:325–331.
  • Chapman MJ, Ginsberg HN, Amarenco P, et al. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: evidence and guidance for management. Eur Heart J. 2011;32:1345–1361.
  • Jenkins AJ, Toth PP, Lyons TJ. SpringerLink (Online service). Lipoproteins in Diabetes Mellitus. Available from: http://dx.doi.org/10.1007/978-1-4614-7554-5.
  • Krauss RM. Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care. 2004;27:1496–1504.
  • Goldberg IJ. Clinical review 124: diabetic dyslipidemia: causes and consequences. J Clin Endocrinol Metab. 2001;86:965–971.
  • Fisher EA, Ginsberg HN. Complexity in the secretory pathway: the assembly and secretion of apolipoprotein B-containing lipoproteins. J Biol Chem. 2002;277:17377–17380.
  • Haas ME, Attie AD, Biddinger SB. The regulation of ApoB metabolism by insulin. Trends Endocrinol Metab. 2013;24:391–397.
  • Wang H, Eckel RH. Lipoprotein lipase: from gene to obesity. Am J Physiol Endocrinol Metab. 2009;297:E271–E288.
  • Calabresi L, Nilsson P, Pinotti E, et al. A novel homozygous mutation in CETP gene as a cause of CETP deficiency in a Caucasian kindred. Atherosclerosis. 2009;205:506–511.
  • Horowitz BS, Goldberg IJ, Merab J, et al. Increased plasma and renal clearance of an exchangeable pool of apolipoprotein A-I in subjects with low levels of high density lipoprotein cholesterol. J Clin Invest. 1993;91:1743–1752.
  • Verges B. Pathophysiology of diabetic dyslipidaemia: where are we? Diabetologia. 2015;58:886–899.
  • Diffenderfer MR, Lamon-Fava S, Marcovina SM, et al. Distinct metabolism of apolipoproteins (a) and B-100 within plasma lipoprotein(a). Metab Clin Exp. 2016;65:381–390.
  • Catapano AL, Graham I, De Backer G, et al. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Eur Heart J. 2016;37:2999–3058.
  • Lawes CM, Vander Hoorn S, Rodgers A. International Society of H. Global burden of blood-pressure-related disease, 2001. Lancet. 2008;371:1513–1518.
  • Lewington S, Clarke R, Qizilbash N, Prospective Studies C, et al. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet. 2002;360:1903–1913.
  • Mancia G, Fagard R, Narkiewicz K, et al. 2013 ESH/ESC Guidelines for the management of arterial hypertension: the Task Force for the management of arterial hypertension of the European Society of Hypertension (ESH) and of the European Society of Cardiology (ESC). J Hypertens. 2013;31:1281–1357.
  • Emdin CA, Rahimi K, Neal B, et al. Blood pressure lowering in type 2 diabetes: a systematic review and meta-analysis. JAMA. 2015;313:603–615.
  • Group SR, Wright JT Jr, Williamson JD, et al. A randomized trial of intensive versus standard blood-pressure control. N Engl J Med. 2015;373:2103–2116.
  • Mancia G, Kjeldsen SE, Zappe DH, et al. Cardiovascular outcomes at different on-treatment blood pressures in the hypertensive patients of the VALUE trial. Eur Heart J. 2016;37:955–964.
  • Bohm M, Schumacher H, Teo KK, et al. Achieved blood pressure and cardiovascular outcomes in high-risk patients: results from ONTARGET and TRANSCEND trials. Lancet. 2017;389:2226–2237.
  • Antza C, Stabouli S, Natsis M, et al. Obesity-induced hypertension: new insights. Curr Pharm Des. 2017. doi:10.2174/1381612823666170608083343
  • Reaven GM, Olefsky J, Farquhar JW. Does hyperglycaemia or hyperinsulinaemia characterise the patient with chemical diabetes? Lancet. 1972;1:1247–1249.
  • Modan M, Halkin H, Almog S, et al. Hyperinsulinemia. A link between hypertension obesity and glucose intolerance. J Clin Invest. 1985;75:809–817.
  • Calles-Escandon J, Mirza SA, Sobel BE, et al. Induction of hyperinsulinemia combined with hyperglycemia and hypertriglyceridemia increases plasminogen activator inhibitor 1 in blood in normal human subjects. Diabetes. 1998;47:290–293.
  • Meigs JB, Mittleman MA, Nathan DM, et al. Hyperinsulinemia, hyperglycemia, and impaired hemostasis: the Framingham Offspring Study. JAMA. 2000;283:221–228.
  • Dhawan J, Bray CL, Warburton R, et al. Insulin resistance, high prevalence of diabetes, and cardiovascular risk in immigrant Asians. Genetic or environmental effect? Br Heart J. 1994;72:413–421.
  • McLaughlin T, Abbasi F, Cheal K, et al. Use of metabolic markers to identify overweight individuals who are insulin resistant. Ann Intern Med. 2003;139:802–809.
  • Ford ES, Abbasi F, Reaven GM. Prevalence of insulin resistance and the metabolic syndrome with alternative definitions of impaired fasting glucose. Atherosclerosis. 2005;181:143–148.
  • Grundy SM. Pre-diabetes, metabolic syndrome, and cardiovascular risk. J Am Coll Cardiol. 2012;59:635–643.
  • Via MA, Mechanick JI. Nutrition in type 2 diabetes and the metabolic syndrome. Med Clin North Am. 2016;100:1285–1302.
  • Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346:393–403.
  • Cicero AF, Colletti A. Role of phytochemicals in the management of metabolic syndrome. Phytomedicine. 2016;23:1134–1144.
  • Westerterp-Plantenga MS, Nieuwenhuizen A, Tome D, et al. Dietary protein, weight loss, and weight maintenance. Annu Rev Nutr. 2009;29:21–41.
  • Layman DK, Clifton P, Gannon MC, et al. Protein in optimal health: heart disease and type 2 diabetes. Am J Clin Nutr. 2008;87:1571S–1575S.
  • Pan A, Sun Q, Bernstein AM, et al. Changes in red meat consumption and subsequent risk of type 2 diabetes mellitus: three cohorts of US men and women. JAMA Intern Med. 2013;173:1328–1335.
  • Virtanen HEK, Koskinen TT, Voutilainen S, et al. Intake of different dietary proteins and risk of type 2 diabetes in men: the Kuopio Ischaemic Heart Disease Risk Factor Study. Br J Nutr. 2017;117:882–893.
  • Chalvon-Demersay T, Azzout-Marniche D, Arfsten J, et al. A systematic review of the effects of plant compared with animal protein sources on features of metabolic syndrome. J Nutr. 2017;147:281–292.
  • Arnoldi A, Boschin G, Zanoni C, et al. The health benefits of sweet lupin seed flours and isolated proteins. J Funct Foods. 2015;18:550–563.
  • Sirtori CR, Lovati MR, Manzoni C, et al. Proteins of white lupin seed, a naturally isoflavone-poor legume, reduce cholesterolemia in rats and increase LDL receptor activity in HepG2 cells. J Nutr. 2004;134:18–23.
  • Bertoglio JC, Calvo MA, Hancke JL, et al. Hypoglycemic effect of lupin seed γ-conglutin in experimental animals and healthy human subjects . Fitoterapia. 2011;82:933–938.
  • Marchesi M, Parolini C, Diani E, et al. Hypolipidaemic and anti-atherosclerotic effects of lupin proteins in a rabbit model. Br J Nutr. 2008;100:707–710.
  • Scirica BM, Bhatt DL, Braunwald E, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369:1317–1326.
  • Lammi C, Zanoni C, Arnoldi A, et al. Peptides derived from soy and lupin protein as dipeptidyl-peptidase IV inhibitors: in vitro biochemical screening and in silico molecular modeling study. J Agric Food Chem. 2016;64:9601–9606.
  • Bahr M, Fechner A, Kramer J, et al. Lupin protein positively affects plasma LDL cholesterol and LDL:HDL cholesterol ratio in hypercholesterolemic adults after four weeks of supplementation: a randomized, controlled crossover study. Nutr J. 2013;12:107.
  • Bahr M, Fechner A, Kiehntopf M, et al. Consuming a mixed diet enriched with lupin protein beneficially affects plasma lipids in hypercholesterolemic subjects: a randomized controlled trial. Clin Nutr. 2015;34:7–14.
  • Sirtori CR, Triolo M, Bosisio R, et al. Hypocholesterolaemic effects of lupin protein and pea protein/fibre combinations in moderately hypercholesterolaemic individuals. Br J Nutr. 2012;107:1176–1183.
  • Spielmann J, Shukla A, Brandsch C, et al. Dietary lupin protein lowers triglyceride concentrations in liver and plasma in rats by reducing hepatic gene expression of sterol regulatory element-binding protein-1c. Ann Nutr Metab. 2007;51:387–392.
  • Pavanello C, Lammi C, Ruscica M, et al. Effects of a lupin protein concentrate on lipids, blood pressure and insulin resistance in moderately dyslipidaemic patients: a randomised controlled trial. J Funct Foods. 2017;37:8–15.
  • Ferri N, Ruscica M. Erratum to: proprotein convertase subtilisin/kexin type 9 (PCSK9) and metabolic syndrome: insights on insulin resistance, inflammation, and atherogenic dyslipidemia. Endocrine. 2016;54:602.
  • Ferri N, Ruscica M. Proprotein convertase subtilisin/kexin type 9 (PCSK9) and metabolic syndrome: insights on insulin resistance, inflammation, and atherogenic dyslipidemia. Endocrine. 2016;54:588–601.
  • Lammi C, Zanoni C, Aiello G, et al. Lupin peptides modulate the protein-protein interaction of PCSK9 with the low density lipoprotein receptor in HepG2 cells. Sci Rep. 2016;6:29931.
  • Lima-Cabello E, Alche V, Foley RC, et al. Narrow-leafed lupin (Lupinus angustifolius L.) beta-conglutin proteins modulate the insulin signaling pathway as potential type 2 diabetes treatment and inflammatory-related disease amelioration. Mol Nutr Food Res. 2017;61. doi:10.1002/mnfr.201600819
  • Anthony MS, Clarkson TB, Bullock BC, et al. Soy protein versus soy phytoestrogens in the prevention of diet-induced coronary artery atherosclerosis of male cynomolgus monkeys. Arterioscler Thromb Vasc Biol. 1997;17:2524–2531.
  • Sirtori CR, Arnoldi A, Johnson SK. Phytoestrogens: end of a tale? Ann Med. 2005;37:423–438.
  • Fukui K, Tachibana N, Wanezaki S, et al. Isoflavone-free soy protein prepared by column chromatography reduces plasma cholesterol in rats. J Agric Food Chem. 2002;50:5717–5721.
  • Lovati MR, Manzoni C, Canavesi A, et al. Soybean protein diet increases low density lipoprotein receptor activity in mononuclear cells from hypercholesterolemic patients. J Clin Invest. 1987;80:1498–1502.
  • Azadbakht L, Kimiagar M, Mehrabi Y, et al. Soy inclusion in the diet improves features of the metabolic syndrome: a randomized crossover study in postmenopausal women. Am J Clin Nutr. 2007;85:735–741.
  • Desroches S, Mauger JF, Ausman LM, et al. Soy protein favorably affects LDL size independently of isoflavones in hypercholesterolemic men and women. J Nutr. 2004;134:574–579.
  • Maki KC, Butteiger DN, Rains TM, et al. Effects of soy protein on lipoprotein lipids and fecal bile acid excretion in men and women with moderate hypercholesterolemia. J Clin Lipidol. 2010;4:531–542.
  • Dove ER, Mori TA, Chew GT, et al. Lupin and soya reduce glycaemia acutely in type 2 diabetes. Br J Nutr. 2011;106:1045–1051.
  • Lin Y, Bolca S, Vandevijvere S, et al. Plant and animal protein intake and its association with overweight and obesity among the Belgian population. Br J Nutr. 2011;105:1106–1116.
  • Vergnaud AC, Norat T, Mouw T, et al. Macronutrient composition of the diet and prospective weight change in participants of the EPIC-PANACEA study. PLoS One. 2013;8:e57300.
  • Gunther AL, Remer T, Kroke A, et al. Early protein intake and later obesity risk: which protein sources at which time points throughout infancy and childhood are important for body mass index and body fat percentage at 7 y of age? Am J Clin Nutr. 2007;86:1765–1772.
  • Ruscica M, Pavanello C, Gandini S, et al. Effect of soy on metabolic syndrome and cardiovascular risk factors: a randomized controlled trial. Eur J Nutr. 2016. doi:10.1007/s00394-016-1333-7
  • Mortensen LS, Hartvigsen ML, Brader LJ, et al. Differential effects of protein quality on postprandial lipemia in response to a fat-rich meal in type 2 diabetes: comparison of whey, casein, gluten, and cod protein. Am J Clin Nutr. 2009;90:41–48.
  • Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell. 2016;164:337–340.
  • Schroeder BO, Backhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nat Med. 2016;22:1079–1089.
  • Rios-Covian D, Salazar N, Gueimonde M, et al. Shaping the metabolism of intestinal bacteroides population through diet to improve human health. Front Microbiol. 2017;8:376.
  • Saad MJ, Santos A, Prada PO. Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology (Bethesda). 2016;31:283–293.
  • Pedersen HK, Gudmundsdottir V, Nielsen HB, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535:376–381.
  • Org E, Blum Y, Kasela S, et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017;18:70.
  • Ussar S, Fujisaka S, Kahn CR. Interactions between host genetics and gut microbiome in diabetes and metabolic syndrome. Mol Metab. 2016;5:795–803.
  • Claesson MJ, Jeffery IB, Conde S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488:178–184.
  • Food JFWWGRoDGftEoPi. Guidelines for the evaluation of probiotics in food. London, Ontario, Canada; 2002.
  • Gibson GR, Probert HM, Loo JV, et al. Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev. 2004;17:259–275.
  • Schrezenmeir J, de Vrese M. Probiotics, prebiotics, and synbiotics-approaching a definition. Am J Clin Nutr. 2001;73:361S–364S.
  • Cho YA, Kim J. Effect of probiotics on blood lipid concentrations: a meta-analysis of randomized controlled trials. Medicine (Baltimore). 2015;94:e1714.
  • Dror T, Dickstein Y, Dubourg G, et al. Microbiota manipulation for weight change. Microb Pathog. 2017;106:146–161.
  • Park S, Bae JH. Probiotics for weight loss: a systematic review and meta-analysis. Nutr Res. 2015;35:566–575.
  • Barengolts E. Gut microbiota, prebiotics, probiotics, and synbiotics in management of obesity and prediabetes: review of randomized controlled trials. Endocr Pract. 2016;22:1224–1234.
  • Sun J, Buys N. Effects of probiotics consumption on lowering lipids and CVD risk factors: a systematic review and meta-analysis of randomized controlled trials. Ann Med. 2015;47:430–440.
  • Zhang Q, Wu Y, Fei X. Effect of probiotics on body weight and body-mass index: a systematic review and meta-analysis of randomized, controlled trials. Int J Food Sci Nutr. 2015;67:571–580.
  • Boutagy NE, McMillan RP, Frisard MI, et al. Metabolic endotoxemia with obesity: is it real and is it relevant? Biochimie. 2016;124:11–20.
  • Luche E, Cousin B, Garidou L, et al. Metabolic endotoxemia directly increases the proliferation of adipocyte precursors at the onset of metabolic diseases through a CD14-dependent mechanism. Mol Metab. 2013;2:281–291.
  • Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031.
  • Flint A, Raben A, Astrup A, et al. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest. 1998;101:515–520.
  • Khalesi S, Sun J, Buys N, et al. Effect of probiotics on blood pressure: a systematic review and meta-analysis of randomized, controlled trials. Hypertension. 2014;64:897–903.
  • Hendijani F, Akbari V. Probiotic supplementation for management of cardiovascular risk factors in adults with type II diabetes: a systematic review and meta-analysis. Clin Nutr. 2017. doi:10.1016/j.clnu.2017.02.015
  • Robles-Vera I, Toral M, Romero M, et al. Antihypertensive effects of probiotics. Curr Hypertens Rep. 2017;19:26.
  • Jauhiainen T, Korpela R. Milk peptides and blood pressure. J Nutr. 2007;137:825S–829S.
  • Sirtori CR, Arnoldi A, Cicero AF. Nutraceuticals for blood pressure control. Ann Med. 2015;47:447–456.
  • Nikbakht E, Khalesi S, Singh I, et al. Effect of probiotics and synbiotics on blood glucose: a systematic review and meta-analysis of controlled trials. Eur J Nutr. 2016. doi:10.1007/s00394-016-1300-3
  • Li C, Li X, Han H, et al. Effect of probiotics on metabolic profiles in type 2 diabetes mellitus: a meta-analysis of randomized, controlled trials. Medicine (Baltimore). 2016;95:e4088.
  • Samah S, Ramasamy K, Lim SM, et al. Probiotics for the management of type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Res Clin Pract. 2016;118:172–182.
  • Akbari V, Hendijani F. Effects of probiotic supplementation in patients with type 2 diabetes: systematic review and meta-analysis. Nutr Rev. 2016;74:774–784.
  • Cani PD, Osto M, Geurts L, et al. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes. 2012;3:279–288.
  • Creely SJ, McTernan PG, Kusminski CM, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab. 2007;292:E740–E747.
  • Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–1772.
  • Shimizu M, Hashiguchi M, Shiga T, et al. Meta-analysis: effects of probiotic supplementation on lipid profiles in normal to mildly hypercholesterolemic individuals. PLoS One. 2015;10:e0139795.
  • Ishimwe N, Daliri EB, Lee BH, et al. The perspective on cholesterol-lowering mechanisms of probiotics. Mol Nutr Food Res. 2015;59:94–105.
  • Kellow NJ, Coughlan MT, Reid CM. Metabolic benefits of dietary prebiotics in human subjects: a systematic review of randomised controlled trials. Br J Nutr. 2014;111(7):1147–1161.
  • Delzenne NM, Cani PD, Daubioul C, et al. Impact of inulin and oligofructose on gastrointestinal peptides. Br J Nutr. 2005;93 Suppl 1:S157–S161.
  • Cani PD, Lecourt E, Dewulf EM, et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr. 2009;90:1236–1243.
  • Beserra BT, Fernandes R, do Rosario VA, et al. A systematic review and meta-analysis of the prebiotics and synbiotics effects on glycaemia, insulin concentrations and lipid parameters in adult patients with overweight or obesity. Clin Nutr. 2015;34:845–858.
  • Khan H, Kunutsor S, Franco OH, et al. Vitamin D, type 2 diabetes and other metabolic outcomes: a systematic review and meta-analysis of prospective studies. Proc Nutr Soc. 2013;72:89–97.
  • Atef SH. Vitamin D assays in clinical laboratory: past, present and future challenges. J Steroid Biochem Mol Biol. 2017. doi:10.1016/j.jsbmb.2017.02.011
  • Holick MF. The vitamin D deficiency pandemic: approaches for diagnosis, treatment and prevention. Rev Endocr Metab Disord. 2017;18:153–165.
  • Holick MF. Vitamin D deficiency. N Engl J Med. 2007;357:266–281.
  • Ross AC, Manson JE, Abrams SA, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab. 2011;96:53–58.
  • Ju SY, Jeong HS, Kim DH. Blood vitamin D status and metabolic syndrome in the general adult population: a dose–response meta-analysis. J Clin Endocrinol Metab. 2014;99:1053–1063.
  • Pereira-Santos M, Costa PR, Assis AM, et al. Obesity and vitamin D deficiency: a systematic review and meta-analysis. Obes Rev. 2015;16:341–349.
  • Slusher AL, McAllister MJ, Huang CJ. A therapeutic role for vitamin D on obesity-associated inflammation and weight-loss intervention. Inflamm Res. 2015;64:565–575.
  • Jorde R. RCTS are the only appropriate way to demonstrate the role of vitamin D in health. J Steroid Biochem Mol Biol. 2017. doi:10.1016/j.jsbmb.2017.05.004
  • Song Y, Wang L, Pittas AG, et al. Blood 25-hydroxy vitamin D levels and incident type 2 diabetes: a meta-analysis of prospective studies. Diabetes Care. 2013;36:1422–1428.
  • Angellotti E, Pittas AG. The role of vitamin D in the prevention of type 2 diabetes. To D or not to D? Endocrinology. 2017. doi:10.1210/en.2017-00265
  • Krause R, Buhring M, Hopfenmuller W, et al. Ultraviolet B and blood pressure. Lancet. 1998;352:709–710.
  • Scragg R, Sowers M, Bell C. Serum 25-hydroxyvitamin D, ethnicity, and blood pressure in the Third National Health and Nutrition Examination Survey. Am J Hypertens. 2007;20:713–719.
  • Jorde R, Figenschau Y, Emaus N, et al. Serum 25-hydroxyvitamin D levels are strongly related to systolic blood pressure but do not predict future hypertension. Hypertension. 2010;55:792–798.
  • Kunutsor SK, Apekey TA, Steur M. Vitamin D and risk of future hypertension: meta-analysis of 283,537 participants. Eur J Epidemiol. 2013;28:205–221.
  • Forman JP, Giovannucci E, Holmes MD, et al. Plasma 25-hydroxyvitamin D levels and risk of incident hypertension. Hypertension. 2007;49:1063–1069.
  • Lee JH, O'Keefe JH, Bell D, et al. Vitamin D deficiency an important, common, and easily treatable cardiovascular risk factor? J Am Coll Cardiol. 2008;52:1949–1956.
  • Sugden JA, Davies JI, Witham MD, et al. Vitamin D improves endothelial function in patients with type 2 diabetes mellitus and low vitamin D levels. Diabet Med. 2008;25:320–325.
  • Beveridge LA, Struthers AD, Khan F, et al. Effect of vitamin D supplementation on blood pressure: a systematic review and meta-analysis incorporating individual patient data. JAMA Intern Med. 2015;175:745–754.
  • Salekzamani S, Bavil AS, Mehralizadeh H, et al. The effects of vitamin D supplementation on proatherogenic inflammatory markers and carotid intima media thickness in subjects with metabolic syndrome: a randomized double-blind placebo-controlled clinical trial. Endocrine. 2017;57:51–59.
  • Momtazi AA, Derosa G, Maffioli P, et al. Role of microRNAs in the therapeutic effects of curcumin in non-cancer diseases. Mol Diagn Ther. 2016;20:335–345.
  • Gordon ON, Luis PB, Sintim HO, et al. Unraveling curcumin degradation: autoxidation proceeds through spiroepoxide and vinylether intermediates en route to the main bicyclopentadione. J Biol Chem. 2015;290:4817–4828.
  • Panahi Y, Khalili N, Sahebi E, et al. Antioxidant effects of curcuminoids in patients with type 2 diabetes mellitus: a randomized controlled trial. Inflammopharmacology. 2017;25:25–31.
  • Panahi Y, Hosseini MS, Khalili N, et al. Effects of supplementation with curcumin on serum adipokine concentrations: a randomized controlled trial. Nutrition. 2016;32:1116–1122.
  • Wongcharoen W, Phrommintikul A. The protective role of curcumin in cardiovascular diseases. Int J Cardiol. 2009;133:145–151.
  • Momtazi AA, Shahabipour F, Khatibi S, et al. Curcumin as a microRNA regulator in cancer: a review. Rev Physiol Biochem Pharmacol. 2016;171:1–38.
  • Sahebkar A. Are curcuminoids effective C-reactive protein-lowering agents in clinical practice? Evidence from a meta-analysis. Phytother Res. 2014;28:633–642.
  • Nakayama H, Tsuge N, Sawada H, et al. A single consumption of curry improved postprandial endothelial function in healthy male subjects: a randomized, controlled crossover trial. Nutr J. 2014;13:67.
  • Sahebkar A. Why it is necessary to translate curcumin into clinical practice for the prevention and treatment of metabolic syndrome? Biofactors. 2013;39:197–208.
  • Seo KI, Choi MS, Jung UJ, et al. Effect of curcumin supplementation on blood glucose, plasma insulin, and glucose homeostasis related enzyme activities in diabetic db/db mice. Mol Nutr Food Res. 2008;52:995–1004.
  • Pari L, Murugan P. Changes in glycoprotein components in streptozotocin–nicotinamide induced type 2 diabetes: influence of tetrahydrocurcumin from Curcuma longa. Plant Foods Hum Nutr. 2007;62:25–29.
  • Chuengsamarn S, Rattanamongkolgul S, Luechapudiporn R, et al. Curcumin extract for prevention of type 2 diabetes. Diabetes Care. 2012;35:2121–2127.
  • Yang YS, Su YF, Yang HW, et al. Lipid-lowering effects of curcumin in patients with metabolic syndrome: a randomized, double-blind, placebo-controlled trial. Phytother Res. 2014;28:1770–1777.
  • Panahi Y, Hosseini MS, Khalili N, et al. Antioxidant and anti-inflammatory effects of curcuminoid-piperine combination in subjects with metabolic syndrome: a randomized controlled trial and an updated meta-analysis. Clin Nutr. 2015;34:1101–1108.
  • Lin XL, Liu MH, Hu HJ, et al. Curcumin enhanced cholesterol efflux by upregulating ABCA1 expression through AMPK-SIRT1-LXRalpha signaling in THP-1 macrophage-derived foam cells. DNA Cell Biol. 2015;34:561–572.
  • Shin SK, Ha TY, McGregor RA, et al. Long-term curcumin administration protects against atherosclerosis via hepatic regulation of lipoprotein cholesterol metabolism. Mol Nutr Food Res. 2011;55:1829–1840.
  • Kumar P, Malhotra P, Ma K, et al. SREBP2 mediates the modulation of intestinal NPC1L1 expression by curcumin. Am J Physiol Gastrointest Liver Physiol. 2011;301:G148–G155.
  • Tai MH, Chen PK, Chen PY, et al. Curcumin enhances cell-surface LDLR level and promotes LDL uptake through downregulation of PCSK9 gene expression in HepG2 cells. Mol Nutr Food Res. 2014;58:2133–2145.
  • Momtazi AA, Banach M, Pirro M, et al. Regulation of PCSK9 by nutraceuticals. Pharmacol Res. 2017;120:157–169.
  • Sahebkar A. Curcuminoids for the management of hypertriglyceridaemia. Nat Rev Cardiol. 2014;11:123.
  • Jager R, Lowery RP, Calvanese AV, et al. Comparative absorption of curcumin formulations. Nutr J. 2014;13:11.
  • Vashisht M, Rani P, Onteru SK, et al. Curcumin encapsulated in milk exosomes resists human digestion and possesses enhanced intestinal permeability in vitro. Appl Biochem Biotechnol. 2017. doi:10.1007/s12010-017-2478-4
  • Panahi Y, Khalili N, Hosseini MS, et al. Lipid-modifying effects of adjunctive therapy with curcuminoids-piperine combination in patients with metabolic syndrome: results of a randomized controlled trial. Complement Ther Med. 2014;22:851–857.
  • Baum L, Cheung SK, Mok VC, et al. Curcumin effects on blood lipid profile in a 6-month human study. Pharmacol Res. 2007;56:509–514.
  • Kong W, Wei J, Abidi P, et al. Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat Med. 2004;10:1344–1351.
  • Lee S, Lim HJ, Park JH, et al. Berberine-induced LDLR up-regulation involves JNK pathway. Biochem Biophys Res Commun. 2007;362:853–857.
  • Caliceti C, Rizzo P, Ferrari R, et al. Novel role of the nutraceutical bioactive compound berberine in lectin-like OxLDL receptor 1-mediated endothelial dysfunction in comparison to lovastatin. Nutr Metab Cardiovasc Dis. 2017;27:552–563.
  • Yin J, Xing H, Ye J. Efficacy of berberine in patients with type 2 diabetes mellitus. Metabolism. 2008;57:712–717.
  • Huang C, Zhang Y, Gong Z, et al. Berberine inhibits 3T3-L1 adipocyte differentiation through the PPARgamma pathway. Biochem Biophys Res Commun. 2006;348:571–578.
  • Ferri N, Corsini A, Sirtori C, et al. PPAR-alpha agonists are still on the rise: an update on clinical and experimental findings. Expert Opin Investig Drugs. 2017;26:593–602.
  • Zhang Y, Li X, Zou D, et al. Treatment of type 2 diabetes and dyslipidemia with the natural plant alkaloid berberine. J Clin Endocrinol Metab. 2008;93:2559–2565.
  • Lee YS, Kim WS, Kim KH, et al. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Diabetes. 2006;55:2256–2264.
  • Yang G, Li X, Li X, et al. Traditional Chinese medicine in cancer care: a review of case series published in the Chinese literature. Evid Based Complement Alternat Med. 2012;2012:751046.
  • Zhou L, Wang X, Yang Y, et al. Berberine attenuates cAMP-induced lipolysis via reducing the inhibition of phosphodiesterase in 3T3-L1 adipocytes. Biochim Biophys Acta. 2011;1812:527–535.
  • Hawley SA, Gadalla AE, Olsen GS, et al. The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism. Diabetes. 2002;51:2420–2425.
  • Foretz M, Hebrard S, Leclerc J, et al. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 2010;120:2355–2369.
  • Zhang DW, Lagace TA, Garuti R, et al. Binding of proprotein convertase subtilisin/kexin type 9 to epidermal growth factor-like repeat A of low density lipoprotein receptor decreases receptor recycling and increases degradation. J Biol Chem. 2007;282:18602–18612.
  • Ferri N, Corsini A, Macchi C, et al. Proprotein convertase subtilisin kexin type 9 and high-density lipoprotein metabolism: experimental animal models and clinical evidence. Transl Res. 2016;173:19–29.
  • Cameron J, Ranheim T, Kulseth MA, et al. Berberine decreases PCSK9 expression in HepG2 cells. Atherosclerosis. 2008;201:266–273.
  • Dong B, Li H, Singh AB, et al. Inhibition of PCSK9 transcription by berberine involves down-regulation of hepatic HNF1alpha protein expression through the ubiquitin–proteasome degradation pathway. J Biol Chem. 2015;290:4047–4058.
  • Ruscica M, Ricci C, Macchi C, et al. Suppressor of cytokine signaling-3 (SOCS-3) induces Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) expression in hepatic HepG2 cell line. J Biol Chem. 2016;291:3508–3519.
  • Briand F, Thieblemont Q, Muzotte E, et al. Upregulating reverse cholesterol transport with cholesteryl ester transfer protein inhibition requires combination with the LDL-lowering drug berberine in dyslipidemic hamsters. Arterioscler Thromb Vasc Biol. 2013;33:13–23.
  • Caliceti C, Franco P, Spinozzi S, et al. Berberine: new insights from pharmacological aspects to clinical evidences in the management of metabolic disorders. CMC. 2016;23:1460–1476.
  • Maeng HJ, Yoo HJ, Kim IW, et al. P-glycoprotein-mediated transport of berberine across Caco-2 cell monolayers. J Pharm Sci. 2002;91:2614–2621.
  • Derosa G, Romano D, D'Angelo A, et al. Berberis aristata combined with Silybum marianum on lipid profile in patients not tolerating statins at high doses. Atherosclerosis. 2015;239:87–92.
  • DeBose-Boyd RA. Feedback regulation of cholesterol synthesis: sterol-accelerated ubiquitination and degradation of HMG CoA reductase. Cell Res. 2008;18:609–621.
  • Childress L, Gay A, Zargar A, et al. Review of red yeast rice content and current Food and Drug Administration oversight. J Clin Lipidol. 2013;7:117–122.
  • Scientific Opinion on the substantiation of health claims related to monacolin K from red yeast rice and maintenance of normal blood LDL cholesterol concentrations (ID 1648, 1700) pursuant to Article 13(1) of Regulation (EC) No. 1924/2006. 2011.
  • Zhao SP, Liu L, Cheng YC, et al. Xuezhikang, an extract of cholestin, protects endothelial function through antiinflammatory and lipid-lowering mechanisms in patients with coronary heart disease. Circulation. 2004;110:915–920.
  • Cicero AF, Morbini M, Rosticci M, et al. Middle-term dietary supplementation with red yeast rice plus coenzyme Q10 improves lipid pattern, endothelial reactivity and arterial stiffness in moderately hypercholesterolemic subjects. Ann Nutr Metab. 2016;68:213–219.
  • Cicero AF, Derosa G, Parini A, et al. Red yeast rice improves lipid pattern, high-sensitivity C-reactive protein, and vascular remodeling parameters in moderately hypercholesterolemic Italian subjects. Nutr Res. 2013;33:622–628.
  • Zhu XY, Li P, Yang YB, et al. Xuezhikang, extract of red yeast rice, improved abnormal hemorheology, suppressed caveolin-1 and increased eNOS expression in atherosclerotic rats. PLoS One. 2013;8:e62731.
  • Verhoeven V, Van der Auwera A, Van Gaal L, et al. Can red yeast rice and olive extract improve lipid profile and cardiovascular risk in metabolic syndrome?: a double blind, placebo controlled randomized trial. BMC Complement Altern Med. 2015;15:52.
  • Xiong X, Wang P, Li X, et al. The effects of red yeast rice dietary supplement on blood pressure, lipid profile, and C-reactive protein in hypertension: a systematic review. Crit Rev Food Sci Nutr. 2017;57:1831–1851.
  • Pirro M, Mannarino MR, Bianconi V, et al. The effects of a nutraceutical combination on plasma lipids and glucose: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2016;110:76–88.
  • Arnaboldi F, Busnelli M, Cornaghi L, et al. High-density lipoprotein deficiency in genetically modified mice deeply affects skin morphology: a structural and ultrastructural study. Exp Cell Res. 2015;338:105–112.
  • Ruscica M, Gomaraschi M, Mombelli G, et al. Nutraceutical approach to moderate cardiometabolic risk: results of a randomized, double-blind and crossover study with Armolipid Plus. J Clin Lipidol. 2014;8:61–68.
  • Affuso F, Ruvolo A, Micillo F, et al. Effects of a nutraceutical combination (berberine, red yeast rice and policosanols) on lipid levels and endothelial function randomized, double-blind, placebo-controlled study. Nutr Metab Cardiovasc Dis. 2010;20:656–661.
  • Lu Z, Kou W, Du B, et al. Effect of Xuezhikang, an extract from red yeast Chinese rice, on coronary events in a Chinese population with previous myocardial infarction. Am J Cardiol. 2008;101:1689–1693.
  • Li JJ, Lu ZL, Kou WR, et al. Impact of Xuezhikang on coronary events in hypertensive patients with previous myocardial infarction from the China Coronary Secondary Prevention Study (CCSPS). Ann Med. 2010;42:231–240.
  • Magni P, Macchi C, Morlotti B, et al. Risk identification and possible countermeasures for muscle adverse effects during statin therapy. Eur J Intern Med. 2015;26:82–88.
  • Cicero AF, Derosa G, Borghi C. Red yeast rice and statin-intolerant patients. Am J Cardiol. 2010;105:1504.
  • Donmez-Altuntas H, Dumlupinar G, Imamoglu N, et al. Effects of the mycotoxin citrinin on micronucleus formation in a cytokinesis-block genotoxicity assay in cultured human lymphocytes. J Appl Toxicol. 2007;27:337–341.
  • Pascual-Ahuir A, Vanacloig-Pedros E, Proft M. Toxicity mechanisms of the food contaminant citrinin: application of a quantitative yeast model. Nutrients. 2014;6:2077–2087.
  • Cicero AF, Tartagni E, Ertek S. Nutraceuticals for metabolic syndrome management: from laboratory to benchside. CVP. 2014;12:565–571.
  • Perez-Martinez P, Mikhailidis DP, Athyros VG, et al. Lifestyle recommendations for the prevention and management of metabolic syndrome: an international panel recommendation. Nutr Rev. 2017;75:307–326.
  • Pirillo A, Catapano AL. Berberine, a plant alkaloid with lipid- and glucose-lowering properties: from in vitro evidence to clinical studies. Atherosclerosis. 2015;243:449–461.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.