ClickCease
+1-915-850-0900 spinedoctors@gmail.com
Select Page

Basal Metabolic Index (BMI)

Back Clinic Basal Metabolic Index (BMI) Functional Medicine and Fitness Team. BMI or Body Mass Index is a statistical measure that compares a person’s height and weight to determine their overall body composition and fat. If the BMI classification is beyond normal, it is considered a progressively increasing risk of cardiovascular disease. Although BMI doesn’t measure body fat directly, it uses weight and height to determine whether an individual is classified as underweight, normal weight, overweight or obese.

BMI is measured by dividing your weight in pounds by the square of your height in inches, then multiplying by 703. The equation looks like this: BMI = (weight / height x height) x 703.

For example if an individual is 125 pounds and 5 feet 4 inches, then the BMI = (125 / 64 x 64) x 703 = 21.4. This BMI puts the individual in the normal weight range.

This measurement correlates moderately well with other body fat measurements, such as skinfold measurements and underwater weighing. This is according to the Centers for Disease Control and Prevention.


Dietary Strategies: Treatment Of Metabolic Syndrome

Dietary Strategies: Treatment Of Metabolic Syndrome

Dietary Strategies:

Abstract: Metabolic syndrome (MetS) is established as the combination of central obesity and different metabolic disturbances, such as insulin resistance, hypertension and dyslipidemia. This cluster of factors affects approximately 10%�50% of adults worldwide and the prevalence has been increasing in epidemic proportions over the last years. Thus, dietary strategies to treat this heterogenic disease are under continuous study. In this sense, diets based on negative-energy-balance, the Mediterranean dietary pattern, n-3 fatty acids, total antioxidant capacity and meal frequency have been suggested as effective approaches to treat MetS. Furthermore, the type and percentage of carbohydrates, the glycemic index or glycemic load, and dietary fiber content are some of the most relevant aspects related to insulin resistance and impaired glucose tolerance, which are important co-morbidities of MetS. Finally, new studies focused on the molecular action of specific nutritional bioactive compounds with positive effects on the MetS are currently an objective of scientific research worldwide. The present review summarizes some of the most relevant dietary approaches and bioactive compounds employed in the treatment of the MetS to date.

Keywords: metabolic syndrome; dietary strategies; bioactive compounds

1. Metabolic Syndrome

dietary healthy unhealthy foodIt was during the period between 1910 and 1920 when it was suggested for the first time that a cluster of associated metabolic disturbances tended to coexist together [1]. Since then, different health organisms have suggested diverse definitions for metabolic syndrome (MetS) but there has not yet been a well-established consensus. The most common definitions are summarized in Table 1. What is clear for all of these is that the MetS is a clinical entity of substantial heterogeneity, commonly represented by the combination of obesity (especially abdominal obesity), hyperglycemia, dyslipidemia and/or hypertension [2�6].

dietary table 1

Obesity consists of an abnormal or excessive fat accumulation, for which the main cause is a chronic imbalance between energy intake and energy expenditure [7,8]. The excess of energy consumed is primarily deposited in the adipose tissue as triglycerides (TG) [9].

Dyslipidemia encompasses elevated serum TG levels, increased low density lipoprotein- cholesterol (LDL-c) particles, and reduced levels of high density lipoprotein-cholesterol (HDL-c) [10]. It is associated with hepatic steatosis [11], dysfunction of pancreatic ?-cells [12] and elevated risk of atherosclerosis [13], among others.

Another main modifiable MetS manifestation is hypertension, which is mainly defined as a resting systolic blood pressure (SBP) ? 140 mmHg or diastolic blood pressure (DBP) ? 90 mmHg or drug prescription to lower hypertension [14]. It usually involves narrowed arteries and is identified as a major cardiovascular and renal risk factor, related to heart and vascular disease, stroke and myocardial infarction [13,15�17].

Hyperglycemia, related insulin resistance and type 2 diabetes mellitus are characterized by an impaired uptake of glucose by the cells, that lead to elevated plasma glucose levels, glycosuria and ketoacidosis [18]. It is responsible for different tissue damage that shortens the life expectancy of diabetics, involving cardiovascular diseases (CVD), atherosclerosis, hypertension [19], ?-cell dysfunction [12], kidney disease [20] or blindness [21]. Currently, diabetes is considered the leading cause of death in developed countries [22].

Moreover, oxidative stress and low grade inflammation are two important mechanisms implicated in the etiology, pathogenesis, and development of MetS [23]. Oxidative stress is defined as an imbalance between the pro-oxidants and antioxidants in the body [24]. It plays a key role in the development of atherosclerosis by different mechanisms such as the oxidation of LDL-c particles [25] or impairment of HDL-c functions [26]. Inflammation is an immune system response to injury hypothesized to be a major mechanism in the pathogenesis and progression of obesity related disorders and the link between adiposity, insulin resistance, MetS and CVD [27].

Although the prevalence of the MetS varies broadly around the word and depends on the source used for its definition, it is clear that over the last 40�50 years the number of people presenting with this syndrome has risen in epidemic proportions [28]. Moreover, the frequency of this syndrome is increased in developed countries, sedentary people, smokers, low socioeconomic status population, as well as in individuals with unhealthy dietary habits [29,30].

For all of this, there is currently a wide concern to find effective strategies to detect, treat and control the comorbidities associated with MetS. This is a complex challenge as MetS is a clinical entity of substantial heterogeneity and therefore, the different cornerstones implicated in its development should be addressed. In this review we compiled and examined different dietary patterns and bioactive compounds that have pointed out to be effective in MetS treatment.

2. Dietary Patterns

dietarySeveral dietary strategies and their potential positive effects on the prevention and treatment of the different metabolic complications associated to the MetS, are described below and summarized in Table 2.

dietary table 22.1. Energy-Restricted Diet Strategies

dietary

Energy restricted diets are probably the most commonly used and studied dietary strategies for combating excess weight and related comorbidities. They consist in personalized regimes that supply less calories than the total energy expended by a specific individual [31].

A hypocaloric diet results in a negative energy balance and subsequently, in body weight reduction [31]. Weight loss is achieved via fat mobilization from different body compartments as a consequence of the lipolysis process necessary to provide energy substrate [32,33]. In people who are overweight or suffering from obesity, as is the case of most people with MetS, weight loss is important as it is associated with improvement of related disorders such as abdominal obesity (visceral adipose tissue), type 2 diabetes, CVD or inflammation [32�36].

Moreover, as described above, low grade inflammation is associated with MetS and obesity. Therefore, of particular importance is the fact that in obese individuals following a hypocaloric diet, a depletion of plasma inflammatory markers such as interleukin (IL)-6 has been observed [34]. Thus, caloric restriction in obese people suffering MetS may improve the whole-body pro-inflammatory state.

At the same time, body weight reduction is associated with improvements in cellular insulin signal transduction, increments in peripheral insulin sensitivity and higher robustness in insulin secretory responses [32,36]. People with excess body weight who are at risk of developing type 2 diabetes, may benefit from a hypocaloric regime by improving plasma glucose levels and insulin resistance.

In addition, different intervention trials have reported a relationship between energy restricted diets and lower risk of developing CVD. In this sense, in studies with obese people following a hypocaloric diet, improvements in lipid profile variables such as reductions of LDL-c and plasma�TG levels, as well as improvements in hypertension via depletion of SBP and DBP levels have been observed [35,37].

Among the different nutritional intervention trials, a reduction of 500�600 kcal a day of the energy requirements is a well-established hypocaloric dietary strategy, which has demonstrated to be effective in weight reduction [38,39]. However, the challenge lies in maintaining the weight loss over time, as many subjects can follow a prescribed diet for a few months, but most people have difficulty in maintaining the acquired habits over the long term [40,41].

2.2. Diets Rich in Omega-3 Fatty Acids

dietary foods omega 3 infographicThe very long-chain eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are essential omega-3 polyunsaturated fatty acids (n-3 PUFAs) for human physiology. Their main dietary sources are fish and algal oils and fatty fish, but they can also be synthesized by humans from ?-linolenic acid [40].

There is a moderate body of evidence suggesting that n-3 PUFAs, mainly EPA and DHA, have a positive role in the prevention and treatment of the pathologies associated to MetS [42].

In this context, it has been described that EPA and DHA have the ability to reduce the risk of developing CVD and cardiometabolic abnormalities as well as CVD-related mortality [42]. These beneficial effects are thought to be mainly due to the ability of these essential fatty acids to reduce plasma TG levels [43].

Moreover, different studies have shown that people following an increased n-3 PUFA diet have reduced plasma levels of the pro-inflammatory cytokines IL-6 and tumor necrosis factor-alpha (TNF?), as well as plasma C-reactive protein (CRP) [44]. These effects are probably mediated by resolvins, maresins and protectins, which are EPA and DHA metabolic products with anti-inflammatory properties [44].

There are some studies that have observed an association between n-3 ingestion and improvements or prevention of type 2 diabetes development. However, other studies found opposite results [44]. Thus, it cannot be made any specific affirmation in this respect.

The European Food Safety Authority recommends and intake of 250 mg EPA + DHA a day, in the general healthy population as a primary prevention of CVD [45]. These amounts can be achieved with an ingestion of 1�2 fatty fish meals per week [45].

2.3. Diets Based on Low Glycemic Index/Load

dietary salad unconstructedOver the last ten years, the concern about the quality of the carbohydrates (CHO) consumed has risen [46]. In this context, the glycemic index (GI) is used as a CHO quality measure. It consists in a ranking on a scale from 0 to 100 that classifies carbohydrate-containing foods according to the postprandial glucose response [47]. The higher the index, the more promptly the postprandial serum glucose rises and the more rapid the insulin response. A quick insulin response leads to rapid hypoglycemia, which is suggested to be associated with an increment of the feeling of hunger and to a subsequent higher caloric intake [47]. The glycemic load (GL) is equal to the GI multiplied by the number of grams of CHO in a serving [48].

There is a theory which states that MetS is a consequence of an elevated intake of high GI foods over time, among others unhealthy dietary habits [49]. In this sense, following a diet rich in high GI CHO has been associated with hyperglycemia, insulin resistance, type 2 diabetes, hypertriglyceridemia, CVD, and obesity [47,50,51], abnormalities directly related to MetS.

On the contrary, a low GI diet has been associated with slower absorption of the CHO and subsequently smaller blood glucose fluctuations, which indicate better glycemic control [46]. In patients with type 2 diabetes, diets based on low GI are associated with reductions in glycated hemoglobin (HbA1c) and fructosamine blood levels, two biomarkers used as key monitoring factors in diabetes management [52,53].

For all of this, it is common to find the limitation of CHO at high GI among the advice for MetS treatment [28], in particular with respect to �ready-to-eat processed foods� including sweetened beverages, soft drinks, cookies, cakes, candy, juice drinks, and other foods which contain high amounts of added sugars [54].

2.4. Diets with High Total Antioxidant Capacity

dietary antioxidant foodsDietary total antioxidant capacity (TAC) is an indicator of diet quality defined as the sum of antioxidant activities of the pool of antioxidants present in a food [55]. These antioxidants have the capacity to act as scavengers of free radicals and other reactive species produced in the organisms [56]. Taking into account that oxidative stress is one of the remarkable unfortunate physiological states of MetS, dietary antioxidants are of main interest in the prevention and treatment of this multifactorial disorder [57]. Accordingly, it is well-accepted that diets with a high content of spices, herbs, fruits, vegetables, nuts and chocolate, are associated with a decreased risk of oxidative stress-related diseases development [58�60]. Moreover, several studies have analyzed the effects of dietary TAC in individuals suffering from MetS or related diseases [61,62]. In the Tehran Lipid and Glucose Study it was demonstrated that a high TAC has beneficial effects on metabolic disorders and especially prevents weight and abdominal fat gain [61]. In the same line, research conducted in our institutions also evidenced that beneficial effects on body weight, oxidative stress biomarkers and other MetS features were positively related with higher TAC consumption in patients suffering from MetS [63�65].

In this sense, the World Health Organization (WHO) recommendation for fruit and vegetables consumption (high TAC foods) for the general population is a minimum of 400 g a day [66]. Moreover, cooking with spices is recommended in order to increase the TAC dietary intake and, at the same time, maintain flavor while reducing salt [67].

2.5. Moderate-High Protein Diets

dietary Protein rich FoodsThe macronutrient distribution set in a weight loss dietary plan has commonly been 50%�55% total caloric value from CHO, 15% from proteins and 30% from lipids [57,68]. However, as most people have difficulty in maintaining weight loss achievements over time [69,70], research on increment of protein intake (>20%) at the expense of CHO was carried out [71�77].

Two mechanisms have been proposed to explain the potential beneficial effects of high-moderate protein diets: the increment of diet-induced thermogenesis [73] and the increase of satiety [78]. The increment of the thermogenesis is explained by the synthesis of peptide bonds, production of urea and gluconeogenesis, which are processes with a higher energy requirement than the metabolism of lipids or CHO [75]. An increment of different appetite-control hormones such as insulin, cholecystokinin or glucagon-like peptide 1, may clarify the satiety effect [79].

Other beneficial effects attributed to moderate-high protein diets in the literature are the improvement of glucose homeostasis [80], the possibility of lower blood lipids [81], the reduction of blood pressure [82], the preservation of lean body mass [83] or the lower of cardiometabolic disease risk [84,85]. However, there are other studies that have not found benefits associated to a moderate-high protein diet [76]. This fact may be explained by the different type of proteins and their amino acid composition [80], as well as by the different type of populations included in each study [85]. Therefore, more research in the field is needed in order to make these results consistent.

In any case, when a hypocaloric diet is implemented, it is necessary to slightly increase the amount of proteins. Otherwise it would be difficult to reach the protein energy requirements, established as 0.83 g/kg/day for isocaloric diets and which should probably be at least 1 g/kg/day for energy-restricted diets [86].

2.6. High Meal Frequency Pattern

dietary eating time

The pattern of increasing meal frequency in weight loss and weight control interventions has currently become popular among professionals [87,88]. The idea is to distribute the total daily energy�intake into more frequently and smaller meals. However, there is no strong evidence about the efficacy of this habit yet [89]. While some investigations have found an inverse association between the increment of meals per day and body weight, body mass index (BMI), fat mass percentage or metabolic diseases such as coronary heart disease or type 2 diabetes [71,88,90�92], others have found no association [93�95].

Different mechanisms by which high meal frequency can have a positive effect on weight and metabolism management have been proposed. An increment of energy expenditure was hypothesized; however, the studies carried out in this line have concluded that total energy expenditure does not differ among different meal frequencies [96,97]. Another postulated hypothesis is that the greater the number of meals a day, the higher the fat oxidation, but again no consensus has been achieved [89,98]. An additional suggested mechanism is that increasing meal frequency leads to plasma glucose levels with lower oscillations and reduced insulin secretion which is thought to contribute to a better appetite control. However, these associations have been found in population with overweight or high glucose levels but in normal-weight or normoglycaemic individuals the results are still inconsistent [93,99�101].

2.7. The Mediterranean Diet

dietary Mediterranean DietThe concept of the Mediterranean Diet (MedDiet) was for the first time defined by the scientific Ancel Keys who observed that those countries around the Mediterranean Sea, which had a characteristic diet, had less risk of suffering coronary heart diseases [102,103].

The traditional MedDiet is characterized by a high intake of extra-virgin olive oil and plant foods (fruits, vegetables, cereals, whole grains, legumes, tree nuts, seeds and olives), low intakes of sweets and red meat and moderate consumption of dairy products, fish and red wine [104].

There is a lot of literature supporting the general health benefits of the MedDiet. In this sense, it has been reported that a high adherence to this dietary pattern protects against mortality and morbidity from several causes [105]. Thus, different studies suggested the MedDiet as a successful tool for the prevention and treatment of MetS and related comorbidities [106�108]. Moreover, recent meta-analysis concluded that the MedDiet is associated with less risk of developing type 2 diabetes and with a better glycemic control in people with this metabolic disorder [107,109,110]. Other studies have found a positive correlation between the adherence to a MedDiet pattern and reduced risk of developing CVD [111�114]. In fact, many studies have found a positive association between following a MedDiet and improvements in lipid profile by reduction of total cholesterol, LDL-c and TG, and an increase in HDL-c [111�115]. Finally, different studies also suggest that the MedDiet pattern may be a good strategy for obesity treatment as it has been associated with significant reductions in body weight and waist circumference [108,116,117].

The high amount of fiber which, among other beneficial effects, helps to weight control providing satiety; and the high antioxidants and anti-inflammatory nutrients such as n-3 fatty acids, oleic acid or phenolic compounds, are thought to be the main contributors to the positive effects attributed to the MedDiet [118].

For all these reasons, efforts to maintain the MedDiet pattern in Mediterranean countries and to implement this dietary habits in westernized countries with unhealthy nutritional patterns should be made.

3. Dietary: Single Nutrients and Bioactive Compounds

dietary Nutrition single nutrientNew studies focused on the molecular action of nutritional bioactive compounds with positive effects on MetS are currently an objective of scientific research worldwide with the aim of designing more personalized strategies in the framework of molecular nutrition. Among them, flavonoids and antioxidant vitamins are some of the most studied compounds with different potential benefits such as antioxidant, vasodilatory, anti-atherogenic, antithrombotic, and anti-inflammatory effects [119]. Table 3 summarizes different nutritional bioactive compounds with potential positive effects on MetS, including the possible molecular mechanism of action involved.

dietary table 3

3.1. Ascorbate

dietary AscorbateVitamin C, ascorbic acid or ascorbate is an essential nutrient as human beings cannot synthesize it. It is a water-soluble antioxidant mainly found in fruits, especially citrus (lemon, orange), and vegetables (pepper, kale) [120]. Several beneficial effects have been associated to this vitamin such as antioxidant and anti-inflammatory properties and prevention or treatment of CVD and type 2 diabetes [121�123].

This dietary component produces its antioxidant effect primarily by quenching damaging free radicals and other reactive oxygen and nitrogen species and therefore preventing molecules such as LDL-c from oxidation [122]. It can also regenerate other oxidized antioxidants like tocopherol [124].

Moreover, it has been described that ascorbic acid may reduce inflammation as it is associated with depletion of CRP levels [125]. This is an important outcome to take in consideration in the treatment of MetS sufferers, as they usually present low grade inflammation [27].

Supplementation with vitamin C have also been associated with prevention of CVD by improving the endothelial function [126] and probably by lowering blood pressure [121]. These effects are thought to be exerted by the ability of vitamin C to enhance the endothelial nitric oxide synthase enzyme (eNOS) activity and to reduce HDL-c glycation [127].

Additionally, several studies have attributed to ascorbate supplementation an antidiabetic effect by improving whole body insulin sensitivity and glucose control in people with type 2 diabetes [123]. These antidiabetic properties are thought to be mediated by optimization of the insulin secretory function of the pancreatic islet cells by increasing muscle sodium-dependent vitamin C transporters (SVCTs) [128].

Despite all of this, it should be taken into account that most people reach ascorbic acid requirements (established at 95�110 mg/day in the general population) from diet and do not need supplementation [122,129]. Besides, it should be considered that an excess of vitamin C ingestion leads to the opposite effect and oxidative particles are formed [130,131].

3.2. Hydroxytyrosol

dietary HydroxytyrosolHydroxytyrosol (3,4-dihydroxyphenylethanol) is a phenolic compound mainly found in olives [132].

It is considered the strongest antioxidant of olive oil and one of the main antioxidants in nature [133]. It acts as a powerful scavenger of free radicals, as a radical chain breaker and as metal chelator [134]. It has the ability of inhibiting LDL-c oxidation by macrophages [132]. In this sense, it is the only phenol recognized by the European Food Safety Authority (EFSA) as a protector of blood lipids from oxidative damage [135].

Hydroxytyrosol has also been reported to have anti-inflammatory effects, probably by suppressing cyclooxygenase activity and inducing eNOS expression [136]. Thus, enhancement of olives/olive oil intakes or hydroxytyroxol supplementation in people suffering from MetS may be a good strategy in order to improve inflammatory status.

Another beneficial effect attributed to this phenolic compound is its cardiovascular protective effect. It presents anti-atherogenic properties by decreasing the expression of vascular cell adhesion protein 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) [132,137], which are probably the result of an inactivation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF?B), activator protein 1 (AP-1), GATA transcription factor and nicotinamide adenine dinucleotide phosphate (NAD(P)H) oxidase [138,139]. Hydroxytyrosol also provides antidyslipidemic effects by lowering plasma levels of LDL-c, total cholesterol and TG, and by rising HDL-c [138].

Despite the beneficial effects attributed to hydfroxytyrosol as an antioxidant, for its antiinflamatory properties and as cardiovascular protector, it should be taken into account that most studies focused on this compound have been performed with mixtures of olive phenols, thus a synergic effect cannot be excluded [140].

3.3. Quercetin

dietaryQuercetin is a predominant flavanol naturally present in vegetables, fruits, green tea or red wine. It is commonly found as glycoside forms, where rutin is the most common and important structure found in nature [141].

Many beneficial effects that can contribute to MetS improvement have been attributed to quercetin. Among them, its antioxidant capacity should be highlighted, as it has been reported to inhibit lipid peroxidation and increase antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT) or glutathione peroxidase (GPX) [142].

Moreover, an anti-inflammatory effect mediated via attenuation of tumor necrosis factor ? (TNF-?), NF?B and mitogen-activated protein kinases (MAPK), as well as depletion of IL-6, IL-1?, IL-8 or monocyte chemoattractant protein-1 (MCP-1) gene expression has also been attributed to this polyphenol [143].

As most people with MetS are overweight or obese, the role of quercetin in body weight reduction and obesity prevention has been of special interest. In this sense, it stands out the capacity of quercetin to inhibit adipogenesis through inducing the activation of AMP-activated protein kinase (AMPK) and decreasing the expression of CCAAT-enhancer-binding protein-? (C/EBP?), peroxisome�proliferator-activated receptor gamma (PPAR?), and sterol regulatory element-binding protein 1 (SREBP-1) [141,144].

According to the antidiabetic effects, it is proposed that quercetin may act as an agonist of peroxisome proliferator-activated receptor gamma (PPAR?), and thus improve insulin-stimulated glucose uptake in mature adipocytes [145]. Moreover, quercetin may ameliorate hyperglycemia by inhibiting glucose transporter 2 (GLUT2) and insulin dependent phosphatidylinositol-3-kinase (PI3K) and blocking tyrosine kinase (TK) [142].

Finally, different studies observed that quercetin has the ability to reduce blood pressure [146�148]. However, the mechanisms of action are not clear, since some authors have suggested that quercetin increases eNOS, contributing to inhibition of platelet aggregation and improvement of the endothelial function [146,147], but there are other studies that have not come across these results [148].

3.4. Resveratrol

dietary

Resveratrol (3,5,4?-trihidroxiestilben) is a phenolic compound mainly found in red grapes and derived products (red wine, grape juice) [149]. It has shown antioxidant and anti-inflammatory activities, and cardioprotective, anti-obesity and antidiabetic capacities [150�156].

The antioxidant effects of resveratrol have been reported to be carried out by scavenging of hydroxyl, superoxide, and metal-induced radicals as well as by antioxidant effects in cells producing reactive oxygen species (ROS) [150].

Moreover, it has been reported that the anti-inflammatory effects of resveratrol are mediated by inhibiting NF?B signaling [151]. Furthermore, this polyphenol reduces the expression of proinflammatory cytokines such as interleukin 6 (IL-6), interleukin 8 (IL-8), TNF-?, monocyte chemoattractant protein-1 (MCP-1) and eNOS [152]. In addition, resveratrol inhibits the cyclooxygenase (COX) expression and activity, a pathway involved in the synthesis of proinflammatory lipid mediators [152].

Concerning the effects of resveratrol against development of type 2 diabetes, it has been reported that treatment of diabetes patients with this polyphenol provides significant improvements in the status of multiple clinically relevant biomarkers such as fasting glucose levels, insulin concentrations or glycated hemoglobin and Homeostasis Model Assessment Insulin Resistance (HOMA-IR) [153,154].

Additionally, cardioprotective effects have been attributed to resveratrol. In this sense, it is suggested that resveratrol improves the endothelial function by producing nitric oxide (NO) through increasing the activity and expression of eNOS. This effect is thought to be conducted through activation of nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-1 (Sirt 1) and 5? AMP-activated protein kinase (AMPK) [155]. Besides, resveratrol exerts endothelial protection by stimulation of NF-E2-related factor 2 (Nrf2) [156] and decreasing the expression of adhesion proteins such as ICAM-1 and VCAM-1 [152].

Finally, it has been described that resveratrol may have a role in preventing obesity as it has been related with energy metabolism improvement, increasing lipolysis and reducing lipogenesis [157]. However, more studies are needed in order to corroborate these findings.

3.5. Tocopherol

dietary vitamin e TocopherolTocopherols, also known as vitamin E, are a family of eight fat-soluble phenolic compounds whose main dietary sources are vegetable oils, nuts and seeds [130,158].

For a long time, it has been suggested that vitamin E could prevent different metabolic diseases as a potent antioxidant, acting as scavenger of lipid peroxyl radicals by hydrogen donating [159]. In this sense, it was described that tocopherols inhibit peroxidation of membrane phospholipids and prevent generation of free radicals in cell membranes [160].

Moreover, it has been shown that supplementation with ?-tocopherol or ?-tocopherol, two of the different isoforms of vitamin E, could have an effect on inflammation status by reducing CRP levels [161]. Additionally, inhibition of COX and protein kinase C (PKC) and reduction of cytokines�such as IL-8 or plasminogen activator inhibitor-1 (PAI-1) are other mechanisms that may contribute to these anti-inflammatory effects [162,163].

However, the beneficial effects attributed to this vitamin previously have lately became controversial as different clinical trials have not come across such benefits, but ineffective or even harmful effects have been observed [164]. It has been recently suggested that this may be explained by the fact that vitamin E may lose most of the antioxidant capacity when ingested by human beings through different mechanisms [162].

3.6. Anthocyanins

dietary Anthocyanins

Anthocyanins are water-soluble polyphenolic compounds responsible for the red, blue and purple colors of berries, black currants, black grapes, peaches, cherries, plums, pomegranate, eggplant, black beans, red radishes, red onions, red cabbage, purple corn or purple sweet potatoes [165�167]. Actually, they are the most abundant polyphenols in fruits and vegetables [167]. Moreover, they can also be found in teas, honey, nuts, olive oil, cocoa, and cereals [168].

These compounds have high antioxidant capacity inhibiting or decreasing free radicals by donating or transferring electrons from hydrogen atoms [167].

Regarding clinical studies, it has been shown that these bioactive compounds may prevent type 2 diabetes development by improving insulin sensitivity [169]. The exact mechanisms by which anthocyanins exert their antidiabetic effect are not yet clear, but an enhancement of the glucose uptake by muscle and adipocyte cells in an insulin-independent manner has been suggested [169].

Moreover, it has been shown that anthocyanins may have the capacity to prevent CVD development by improving endothelial function via increasing brachial artery flow-mediated dilation and HDL-c, and decreasing serum VCAM-1 and LDL-c concentrations [170�173].

Finally, these polyphenolic compounds may exert anti-inflamatory effects via reducing proinflamatory molecules such as IL-8, IL-1? or CRP [172,174].

However, most studies have used anthocyanin-rich extracts instead of purified anthocyanins; thus, a synergic effect with other polyphenols cannot be discarded.

3.7. Catechins

dietary tea leaves CatechinsCatechins are polyphenols that can be found in a variety of foods including fruits, vegetables, chocolate, wine, and tea [175]. The epigallocatechin 3-gallate present in tea leaves is the catechin class most studied [176].

Anti-obesity effects have been attributed to these polyphenols in different studies [176]. The mechanisms of action suggested to explain these beneficial effects on body weight are: increasing energy expenditure and fat oxidation, and reducing fat absorption [177]. It is thought that energy expenditure is enhanced by catechol-O-methyltransferase and phosphodiesterase inhibition, which stimulates the sympathetic nervous system causing an activation of the brown adipose tissue [178]. Fat oxidation is mediated by upregulation of acyl-CoA dehydrogenase and peroxisomal b-oxidation enzymes [178,179].

Moreover, catechin intake has also been associated with lower risk of CVD development by improving lipid biomarkers. Thus, it has been reported that consumption of this kind of polyphenols may increase HDL-c and decrease LDL-c and total cholesterol [180].

Finally, and antidiabetic effect has also been related to catechin comsumption, lowering fasting glucose levels [175] and improving insulin sensitivity [178].

4. Conclusions

As the prevalence of MetS reaches epidemic rates, the finding of an effective and easy-to-follow dietary strategy to combat this heterogenic disease is still a pending subject. This work recompiled different dietary nutrients and nutritional patterns with potential benefits in the prevention and�treatment of MetS and related comorbidities (Figure 1) with the aim of facilitating future clinical�studies in this area. The challenge now is to introduce precision bioactive compounds in personalized�nutritional patterns in order to gain the most benefits for prevention and treatment of this disease�through nutrition.

dietary fig 1

Conflicts of Interest: The authors declare no conflict of interest.

blank
References:

1. Sarafidis, P.A.; Nilsson, P.M. The metabolic syndrome: A glance at its history. J. Hypertens. 2006, 24, 621�626.
[CrossRef] [PubMed]
2. Alberti, K.G.; Zimmet, P.Z. Definition, diagnosis and classification of diabetes mellitus and its complications.
Part 1: Diagnosis and classification of diabetes mellitus provisional report of a WHO consultation.
Diabet. Med. 1998, 15, 539�553. [CrossRef]
3. Balkau, B.; Charles, M.A. Comment on the provisional report from the WHO consultation. European Group
for the Study of Insulin Resistance (EGIR). Diabet. Med. 1999, 16, 442�423. [PubMed]
4. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. 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.
5. Grundy, S.M.; Cleeman, J.I.; Daniels, S.R.; Donato, K.A.; Eckel, R.H.; Franklin, B.A.; Gordon, D.J.;
Krauss, R.M.; Savage, P.J.; Smith, S.C., Jr.; et al. Diagnosis and management of the metabolic syndrome:
An American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation
2005, 112, 2735�2752. [CrossRef] [PubMed]
6. Alberti, K.G.; Zimmet, P.; Shaw, J. The metabolic syndrome�A new worldwide definition. Lancet 2005, 366,
1059�1062. [CrossRef]
7. Selassie, M.; Sinha, A.C. The epidemiology and aetiology of obesity: A global challenge. Best Pract. Res.
Clin. Anaesthesiol. 2011, 25, 1�9. [CrossRef] [PubMed]
8. WHO, W.H.O. Available online: www.who.int/mediacentre/factsheets/fs311/es/ (accessed on
4 June 2016).
9. Shimano, H. Novel qualitative aspects of tissue fatty acids related to metabolic regulation: Lessons from
Elovl6 knockout. Prog. Lipid Res. 2012, 51, 267�271. [CrossRef] [PubMed]
10. Bosomworth, N.J. Approach to identifying and managing atherogenic dyslipidemia: A metabolic
consequence of obesity and diabetes. Can. Fam. Phys. 2013, 59, 1169�1180.
11. Vidal-Puig, A. The Metabolic Syndrome and its Complex Pathophysiology. In A Systems Biology Approach to
Study Metabolic Syndrome; Oresic, M., Ed.; Springer: New York, NY, USA, 2014; pp. 3�16.
12. Poitout, V.; Robertson, R.P. Glucolipotoxicity: Fuel excess and beta-cell dysfunction. Endocr. Rev. 2008, 29,
351�366. [CrossRef] [PubMed]
13. Rizza, W.; Veronese, N.; Fontana, L. What are the roles of calorie restriction and diet quality in promoting
healthy longevity? Ageing Res. Rev. 2014, 13, 38�45. [CrossRef] [PubMed]
14. Lloyd-Jones, D.M.; Levy, D. Epidemiology of Hypertension. In Hypertension: A Companion to Braunwald�s
Heart Disease; Black, H.R., Elliott, W.J., Eds.; Elsevier: Philadephia, PA, USA, 2013; pp. 1�11.
15. Zanchetti, A. Challenges in hypertension: Prevalence, definition, mechanisms and management. J. Hypertens.
2014, 32, 451�453. [CrossRef] [PubMed]
16. Thomas, G.; Shishehbor, M.; Brill, D.; Nally, J.V., Jr. New hypertension guidelines: One size fits most?
Clevel. Clin. J. Med. 2014, 81, 178�188. [CrossRef] [PubMed]
17. James, P.A.; Oparil, S.; Carter, B.L.; Cushman, W.C.; Dennison-Himmelfarb, C.; Handler, J.; Lackland, D.T.;
LeFevre, M.L.; MacKenzie, T.D.; Ogedegbe, O.; et al. 2014 evidence-based guideline for the management
of high blood pressure in adults: Report from the panel members appointed to the Eighth Joint National
Committee (JNC 8). JAMA 2014, 311, 507�520. [CrossRef] [PubMed]
18. Klandorf, H.; Chirra, A.R.; DeGruccio, A.; Girman, D.J. Dimethyl sulfoxide modulation of diabetes onset in
NOD mice. Diabetes 1989, 38, 194�197. [CrossRef] [PubMed]
19. Ballard, K.D.; Mah, E.; Guo, Y.; Pei, R.; Volek, J.S.; Bruno, R.S. Low-fat milk ingestion prevents postprandial
hyperglycemia-mediated impairments in vascular endothelial function in obese individuals with metabolic
syndrome. J. Nutr. 2013, 143, 1602�1610. [CrossRef] [PubMed]
20. Pugliese, G.; Solini, A.; Bonora, E.; Orsi, E.; Zerbini, G.; Fondelli, C.; Gruden, G.; Cavalot, F.; Lamacchia, O.;
Trevisan, R.; et al. Distribution of cardiovascular disease and retinopathy in patients with type 2 diabetes
according to different classification systems for chronic kidney disease: A cross-sectional analysis of the
renal insufficiency and cardiovascular events (RIACE) Italian multicenter study. Cardiovasc. Diabetol. 2014,
13, 59. [PubMed]
21. Asif, M. The prevention and control the type-2 diabetes by changing lifestyle and dietary pattern. J. Educ.
Health Promot. 2014, 3, 1. [CrossRef] [PubMed]
22. Russell, W.R.; Baka, A.; Bjorck, I.; Delzenne, N.; Gao, D.; Griffiths, H.R.; Hadjilucas, E.; Juvonen, K.;
Lahtinen, S.; Lansink, M.; et al. Impact of Diet Composition on Blood Glucose Regulation. Crit. Rev. Food
Sci. Nutr. 2016, 56, 541�590. [CrossRef] [PubMed]
23. Soares, R.; Costa, C. Oxidative Stress, Inflammation and Angiogenesis in the Metabolic Syndrome; Springer:
Heidelberg, Germany, 2009.
24. Rahal, A.; Kumar, A.; Singh, V.; Yadav, B.; Tiwari, R.; Chakraborty, S.; Dhama, K. Oxidative Stress,
Prooxidants, and Antioxidants: The Interplay. BioMed Res. Int. 2014, 2014, 761264. [CrossRef] [PubMed]
25. Parthasarathy, S.; Litvinov, D.; Selvarajan, K.; Garelnabi, M. Lipid peroxidation and decomposition�Conflicting
roles in plaque vulnerability and stability. Biochim. Biophys. Acta 2008, 1781, 221�231. [CrossRef] [PubMed]
26. McGrowder, D.; Riley, C.; Morrison, E.Y.; Gordon, L. The role of high-density lipoproteins in reducing the
risk of vascular diseases, neurogenerative disorders, and cancer. Cholesterol 2011, 2011, 496925. [CrossRef]
[PubMed]
27. Ferri, N.; Ruscica, M. Proprotein convertase subtilisin/kexin type 9 (PCSK9) and metabolic syndrome:
Insights on insulin resistance, inflammation, and atherogenic dyslipidemia. Endocrine 2016. [CrossRef]
28. Oresic, M.; Vidal-Puig, A. A Systems Biology Approach to Study Metabolic Syndrome; Springer: Heidelberg,
Germany, 2014.
29. Lee, E.G.; Choi, J.H.; Kim, K.E.; Kim, J.H. Effects of a Walking Program on Self-management and Risk Factors
of Metabolic Syndrome in Older Korean Adults. J. Phys. Ther. Sci. 2014, 26, 105�109. [CrossRef] [PubMed]
30. Bernabe, G.J.; Zafrilla, R.P.; Mulero, C.J.; Gomez, J.P.; Leal, H.M.; Abellan, A.J. Biochemical and nutritional
markers and antioxidant activity in metabolic syndrome. Endocrinol. Nutr. 2013, 61, 302�308.
31. Bales, C.W.; Kraus, W.E. Caloric restriction: Implications for human cardiometabolic health. J. Cardiopulm.
Rehabil. Prev. 2013, 33, 201�208. [CrossRef] [PubMed]
32. Grams, J.; Garvey, W.T. Weight Loss and the Prevention and Treatment of Type 2 Diabetes Using Lifestyle
Therapy, Pharmacotherapy, and Bariatric Surgery: Mechanisms of Action. Curr. Obes. Rep. 2015, 4, 287�302.
[CrossRef] [PubMed]
33. Lazo, M.; Solga, S.F.; Horska, A.; Bonekamp, S.; Diehl, A.M.; Brancati, F.L.; Wagenknecht, L.E.; Pi-Sunyer, F.X.;
Kahn, S.E.; Clark, J.M. Effect of a 12-month intensive lifestyle intervention on hepatic steatosis in adults with
type 2 diabetes. Diabetes Care 2010, 33, 2156�2163. [CrossRef] [PubMed]
34. Rossmeislova, L.; Malisova, L.; Kracmerova, J.; Stich, V. Adaptation of human adipose tissue to hypocaloric
diet. Int. J. Obes. 2013, 37, 640�650. [CrossRef] [PubMed]
35. Wing, R.R.; Lang, W.; Wadden, T.A.; Safford, M.; Knowler, W.C.; Bertoni, A.G.; Hill, J.O.; Brancati, F.L.;
Peters, A.; Wagenknecht, L. Benefits of modest weight loss in improving cardiovascular risk factors in
overweight and obese individuals with type 2 diabetes. Diabetes Care 2011, 34, 1481�1486. [CrossRef]
[PubMed]
36. Golay, A.; Brock, E.; Gabriel, R.; Konrad, T.; Lalic, N.; Laville, M.; Mingrone, G.; Petrie, J.; Phan, T.M.;
Pietilainen, K.H.; et al. Taking small steps towards targets�Perspectives for clinical practice in diabetes,
cardiometabolic disorders and beyond. Int. J. Clin. Pract. 2013, 67, 322�332. [CrossRef] [PubMed]
37. Fock, K.M.; Khoo, J. Diet and exercise in management of obesity and overweight. J. Gastroenterol. Hepatol.
2013, 28, 59�63. [CrossRef] [PubMed]
38. Abete, I.; Parra, D.; Martinez, J.A. Energy-restricted diets based on a distinct food selection affecting the
glycemic index induce different weight loss and oxidative response. Clin. Nutr. 2008, 27, 545�551. [CrossRef]
[PubMed]
39. Alberti, K.G.; Eckel, R.H.; Grundy, S.M.; Zimmet, P.Z.; Cleeman, J.I.; Donato, K.A.; Fruchart, J.C.; James, W.P.;
Loria, C.M.; Smith, S.C., Jr. 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. [PubMed]
40. Fleming, J.A.; Kris-Etherton, P.M. The evidence for alpha-linolenic acid and cardiovascular disease benefits:
Comparisons with eicosapentaenoic acid and docosahexaenoic acid. Adv. Nutr. 2014, 5, 863S�876S. [CrossRef]
[PubMed]
41. Gray, B.; Steyn, F.; Davies, P.S.; Vitetta, L. Omega-3 fatty acids: A review of the effects on adiponectin and
leptin and potential implications for obesity management. Eur. J. Clin. Nutr. 2013, 67, 1234�1242. [CrossRef]
[PubMed]
42. Wen, Y.T.; Dai, J.H.; Gao, Q. Effects of Omega-3 fatty acid on major cardiovascular events and mortality
in patients with coronary heart disease: A meta-analysis of randomized controlled trials. Nutr. Metab.
Cardiovasc. Dis. 2014, 24, 470�475. [CrossRef] [PubMed]
43. Lopez-Huertas, E. The effect of EPA and DHA on metabolic syndrome patients: A systematic review of
randomised controlled trials. Br. J. Nutr. 2012, 107, 185�194. [CrossRef] [PubMed]
44. Maiorino, M.I.; Chiodini, P.; Bellastella, G.; Giugliano, D.; Esposito, K. Sexual dysfunction in women with
cancer: A systematic review with meta-analysis of studies using the Female Sexual Function Index. Endocrine
2016, 54, 329�341. [CrossRef] [PubMed]
45. EFSA NDA Panel (EFSA Panel on Dietetic Products, Nutrition and Allergies). Scientific Opinion on Dietary
Reference Values for fats, including saturated fatty acids, polyunsaturated fatty acids, monounsaturated
fatty acids, trans fatty acids, and cholesterol1. EFSA J. 2010, 8, 1461�1566.
46. Bellastella, G.; Bizzarro, A.; Aitella, E.; Barrasso, M.; Cozzolino, D.; di Martino, S.; Esposito, K.; de Bellis, A.
Pregnancy may favour the development of severe autoimmune central diabetes insipidus in women with
vasopressin cell antibodies: Description of two cases. Eur. J. Endocrinol. 2015, 172, K11�K17. [CrossRef]
[PubMed]
47. Sun, F.H.; Li, C.; Zhang, Y.J.; Wong, S.H.; Wang, L. Effect of Glycemic Index of Breakfast on Energy Intake at
Subsequent Meal among Healthy People: A Meta-Analysis. Nutrients 2016, 8, 37. [CrossRef] [PubMed]
48. Barclay, A.W.; Brand-Miller, J.C.; Wolever, T.M. Glycemic index, glycemic load, and glycemic response are
not the same. Diabetes Care 2005, 28, 1839�1840. [CrossRef] [PubMed]
49. Nakagawa, T.; Hu, H.; Zharikov, S.; Tuttle, K.R.; Short, R.A.; Glushakova, O.; Ouyang, X.; Feig, D.I.;
Block, E.R.; Herrera-Acosta, J.; et al. A causal role for uric acid in fructose-induced metabolic syndrome.
Am. J. Physiol. Ren. Physiol. 2006, 290, F625�F631. [CrossRef] [PubMed]
50. Symons Downs, D.; Hausenblas, H.A. Women�s exercise beliefs and behaviors during their pregnancy and
postpartum. J. Midwifery Women Health 2004, 49, 138�144.
51. Brand-Miller, J.; McMillan-Price, J.; Steinbeck, K.; Caterson, I. Dietary glycemic index: Health implications.
J. Am. Coll. Nutr. 2009, 28, 446S�449S. [CrossRef] [PubMed]
52. Thomas, D.; Elliott, E.J. Low glycaemic index, or low glycaemic load, diets for diabetes mellitus.
Cochrane Database Syst. Rev. 2009. [CrossRef]
53. Barrea, L.; Balato, N.; di Somma, C.; Macchia, P.E.; Napolitano, M.; Savanelli, M.C.; Esposito, K.; Colao, A.;
Savastano, S. Nutrition and psoriasis: Is there any association between the severity of the disease and
adherence to the Mediterranean diet? J. Transl. Med. 2015, 13, 18. [CrossRef] [PubMed]
54. Mathias, K.C.; Ng, S.W.; Popkin, B. Monitoring changes in the nutritional content of ready-to-eat grain-based
dessert products manufactured and purchased between 2005 and 2012. J. Acad. Nutr. Diet. 2015, 115, 360�368.
[CrossRef] [PubMed]
55. Serafini, M.; del Rio, D. Understanding the association between dietary antioxidants, redox status and
disease: Is the Total Antioxidant Capacity the right tool? Redox Rep. 2004, 9, 145�152. [CrossRef] [PubMed]
56. Bellastella, G.; Maiorino, M.I.; Olita, L.; della Volpe, E.; Giugliano, D.; Esposito, K. Premature ejaculation is
associated with glycemic control in Type 1 diabetes. J. Sex. Med. 2015, 12, 93�99. [CrossRef] [PubMed]
57. Zulet, M.A.; Moreno-Aliaga, M.J.; Martinez, J.A. Dietary Determinants of Fat Mass and Body Composition.
In Adipose Tissue Biology; Symonds, M.E., Ed.; Springer: New York, NY, USA, 2012; pp. 271�315.
58. Carlsen, M.H.; Halvorsen, B.L.; Holte, K.; Bohn, S.K.; Dragland, S.; Sampson, L.; Willey, C.; Senoo, H.;
Umezono, Y.; Sanada, C.; et al. The total antioxidant content of more than 3100 foods, beverages, spices,
herbs and supplements used worldwide. Nutr. J. 2010, 9, 3. [CrossRef] [PubMed]
59. Harasym, J.; Oledzki, R. Effect of fruit and vegetable antioxidants on total antioxidant capacity of blood
plasma. Nutrition 2014, 30, 511�517. [CrossRef] [PubMed]
60. Maiorino, M.I.; Bellastella, G.; Petrizzo, M.; della Volpe, E.; Orlando, R.; Giugliano, D.; Esposito, K. Circulating
endothelial progenitor cells in type 1 diabetic patients with erectile dysfunction. Endocrine 2015, 49, 415�421.
[CrossRef] [PubMed]
61. Bahadoran, Z.; Golzarand, M.; Mirmiran, P.; Shiva, N.; Azizi, F. Dietary total antioxidant capacity and the
occurrence of metabolic syndrome and its components after a 3-year follow-up in adults: Tehran Lipid and
Glucose Study. Nutr. Metab. 2012, 9, 70. [CrossRef] [PubMed]
62. Chrysohoou, C.; Esposito, K.; Giugliano, D.; Panagiotakos, D.B. Peripheral Arterial Disease and
Cardiovascular Risk: The Role of Mediterranean Diet. Angiology 2015, 66, 708�710. [CrossRef] [PubMed]
63. De la Iglesia, R.; Lopez-Legarrea, P.; Celada, P.; Sanchez-Muniz, F.J.; Martinez, J.A.; Zulet, M.A. Beneficial
effects of the RESMENA dietary pattern on oxidative stress in patients suffering from metabolic syndrome
with hyperglycemia are associated to dietary TAC and fruit consumption. Int. J. Mol. Sci. 2013, 14, 6903�6919.
[CrossRef] [PubMed]
64. Lopez-Legarrea, P.; de la Iglesia, R.; Abete, I.; Bondia-Pons, I.; Navas-Carretero, S.; Forga, L.; Martinez, J.A.;
Zulet, M.A. Short-term role of the dietary total antioxidant capacity in two hypocaloric regimes on obese
with metabolic syndrome symptoms: The RESMENA randomized controlled trial. Nutr. Metab. 2013, 10, 22.
[CrossRef] [PubMed]
65. Puchau, B.; Zulet, M.A.; de Echavarri, A.G.; Hermsdorff, H.H.; Martinez, J.A. Dietary total antioxidant
capacity is negatively associated with some metabolic syndrome features in healthy young adults. Nutrition
2010, 26, 534�541. [CrossRef] [PubMed]
66. World Health Organization. Obesity: Preventing and Managing the Global Epidemic; Report of a WHO
Consultation; World Health Organization Technical Report Series; WHO: Geneva, Switzerland, 2000.
67. Tapsell, L.C.; Hemphill, I.; Cobiac, L.; Patch, C.S.; Sullivan, D.R.; Fenech, M.; Roodenrys, S.; Keogh, J.B.;
Clifton, P.M.; Williams, P.G.; et al. Health benefits of herbs and spices: The past, the present, the future.
Med. J. Aust. 2006, 185, S4�S24. [PubMed]
68. Abete, I.; Astrup, A.; Martinez, J.A.; Thorsdottir, I.; Zulet, M.A. Obesity and the metabolic syndrome: Role of
different dietary macronutrient distribution patterns and specific nutritional components on weight loss and
maintenance. Nutr. Rev. 2010, 68, 214�231. [CrossRef] [PubMed]
69. Ebbeling, C.B.; Swain, J.F.; Feldman, H.A.; Wong, W.W.; Hachey, D.L.; Garcia-Lago, E.; Ludwig, D.S. Effects
of dietary composition on energy expenditure during weight-loss maintenance. JAMA 2012, 307, 2627�2634.
[CrossRef] [PubMed]
70. Abete, I.; Goyenechea, E.; Zulet, M.A.; Martinez, J.A. Obesity and metabolic syndrome: Potential benefit
from specific nutritional components. Nutr. Metab. Cardiovasc. Dis. 2011, 21, B1�B15. [CrossRef] [PubMed]
71. Arciero, P.J.; Ormsbee, M.J.; Gentile, C.L.; Nindl, B.C.; Brestoff, J.R.; Ruby, M. Increased protein intake and
meal frequency reduces abdominal fat during energy balance and energy deficit. Obesity 2013, 21, 1357�1366.
[CrossRef] [PubMed]
72. Wikarek, T.; Chudek, J.; Owczarek, A.; Olszanecka-Glinianowicz, M. Effect of dietary macronutrients on
postprandial incretin hormone release and satiety in obese and normal-weight women. Br. J. Nutr. 2014, 111,
236�246. [CrossRef] [PubMed]
73. Bray, G.A.; Smith, S.R.; de Jonge, L.; Xie, H.; Rood, J.; Martin, C.K.; Most, M.; Brock, C.; Mancuso, S.;
Redman, L.M. Effect of dietary protein content on weight gain, energy expenditure, and body composition
during overeating: A randomized controlled trial. JAMA 2012, 307, 47�55. [CrossRef] [PubMed]
74. Westerterp-Plantenga, M.S.; Nieuwenhuizen, A.; Tome, D.; Soenen, S.; Westerterp, K.R. Dietary protein,
weight loss, and weight maintenance. Annu. Rev. Nutr. 2009, 29, 21�41. [CrossRef] [PubMed]
75. Koppes, L.L.; Boon, N.; Nooyens, A.C.; van Mechelen, W.; Saris, W.H. Macronutrient distribution over
a period of 23 years in relation to energy intake and body fatness. Br. J. Nutr. 2009, 101, 108�115. [CrossRef]
[PubMed]
76. De Jonge, L.; Bray, G.A.; Smith, S.R.; Ryan, D.H.; de Souza, R.J.; Loria, C.M.; Champagne, C.M.;
Williamson, D.A.; Sacks, F.M. Effect of diet composition and weight loss on resting energy expenditure in
the POUNDS LOST study. Obesity 2012, 20, 2384�2389. [CrossRef] [PubMed]
77. Stocks, T.; Angquist, L.; Hager, J.; Charon, C.; Holst, C.; Martinez, J.A.; Saris, W.H.; Astrup, A.; Sorensen, T.I.;
Larsen, L.H. TFAP2B-dietary protein and glycemic index interactions and weight maintenance after weight
loss in the DiOGenes trial. Hum. Hered. 2013, 75, 213�219. [CrossRef] [PubMed]
78. Giugliano, D.; Maiorino, M.I.; Esposito, K. Linking prediabetes and cancer: A complex issue. Diabetologia
2015, 58, 201�202. [CrossRef] [PubMed]
79. Bendtsen, L.Q.; Lorenzen, J.K.; Bendsen, N.T.; Rasmussen, C.; Astrup, A. Effect of dairy proteins on appetite,
energy expenditure, body weight, and composition: A review of the evidence from controlled clinical trials.
Adv. Nutr. 2013, 4, 418�438. [CrossRef] [PubMed]
80. Heer, M.; Egert, S. Nutrients other than carbohydrates: Their effects on glucose homeostasis in humans.
Diabetes Metab. Res. Rev. 2015, 31, 14�35. [CrossRef] [PubMed]
81. Layman, D.K.; Evans, E.M.; Erickson, D.; Seyler, J.; Weber, J.; Bagshaw, D.; Griel, A.; Psota, T.; Kris-Etherton, P.
A moderate-protein diet produces sustained weight loss and long-term changes in body composition and
blood lipids in obese adults. J. Nutr. 2009, 139, 514�521. [CrossRef] [PubMed]
82. Pedersen, A.N.; Kondrup, J.; Borsheim, E. Health effects of protein intake in healthy adults: A systematic
literature review. Food Nutr. Res. 2013, 57, 21245. [CrossRef] [PubMed]
83. Daly, R.M.; O�Connell, S.L.; Mundell, N.L.; Grimes, C.A.; Dunstan, D.W.; Nowson, C.A. Protein-enriched
diet, with the use of lean red meat, combined with progressive resistance training enhances lean tissue mass
and muscle strength and reduces circulating IL-6 concentrations in elderly women: A cluster randomized
controlled trial. Am. J. Clin. Nutr. 2014, 99, 899�910. [CrossRef] [PubMed]
84. Arciero, P.J.; Gentile, C.L.; Pressman, R.; Everett, M.; Ormsbee, M.J.; Martin, J.; Santamore, J.; Gorman, L.;
Fehling, P.C.; Vukovich, M.D.; et al. Moderate protein intake improves total and regional body composition
and insulin sensitivity in overweight adults. Metab. Clin. Exp. 2008, 57, 757�765. [CrossRef] [PubMed]
85. Gregory, S.M.; Headley, S.A.; Wood, R.J. Effects of dietary macronutrient distribution on vascular integrity in
obesity and metabolic syndrome. Nutr. Rev. 2011, 69, 509�519. [CrossRef] [PubMed]
86. Consenso FESNAD-SEEDO. Recomendaciones nutricionales basadas en la evidencia para la prevenci�n y el
tratamiento del sobrepeso y la obesidad en adultos (Consenso FESNAD-SEEDO). Rev. Esp. Obes. 2011, 10, 36.
87. Jakubowicz, D.; Froy, O.; Wainstein, J.; Boaz, M. Meal timing and composition influence ghrelin levels,
appetite scores and weight loss maintenance in overweight and obese adults. Steroids 2012, 77, 323�331.
[CrossRef] [PubMed]
88. Schwarz, N.A.; Rigby, B.R.; La Bounty, P.; Shelmadine, B.; Bowden, R.G. A review of weight control strategies
and their effects on the regulation of hormonal balance. J. Nutr. Metab. 2011, 2011, 237932. [CrossRef]
[PubMed]
89. Ohkawara, K.; Cornier, M.A.; Kohrt, W.M.; Melanson, E.L. Effects of increased meal frequency on fat
oxidation and perceived hunger. Obesity 2013, 21, 336�343. [CrossRef] [PubMed]
90. Ekmekcioglu, C.; Touitou, Y. Chronobiological aspects of food intake and metabolism and their relevance on
energy balance and weight regulation. Obes. Rev. 2011, 12, 14�25. [CrossRef] [PubMed]
91. Lioret, S.; Touvier, M.; Lafay, L.; Volatier, J.L.; Maire, B. Are eating occasions and their energy content related
to child overweight and socioeconomic status? Obesity 2008, 16, 2518�2523. [CrossRef] [PubMed]
92. Bhutani, S.; Varady, K.A. Nibbling versus feasting: Which meal pattern is better for heart disease prevention?
Nutr. Rev. 2009, 67, 591�598. [CrossRef] [PubMed]
93. Leidy, H.J.; Tang, M.; Armstrong, C.L.; Martin, C.B.; Campbell, W.W. The effects of consuming frequent,
higher protein meals on appetite and satiety during weight loss in overweight/obese men. Obesity 2011, 19,
818�824. [CrossRef] [PubMed]
94. Mills, J.P.; Perry, C.D.; Reicks, M. Eating frequency is associated with energy intake but not obesity in midlife
women. Obesity 2011, 19, 552�559. [CrossRef] [PubMed]
95. Cameron, J.D.; Cyr, M.J.; Doucet, E. Increased meal frequency does not promote greater weight loss in subjects
who were prescribed an 8-week equi-energetic energy-restricted diet. Br. J. Nutr. 2010, 103, 1098�1101.
[CrossRef] [PubMed]
96. Smeets, A.J.; Lejeune, M.P.; Westerterp-Plantenga, M.S. Effects of oral fat perception by modified sham
feeding on energy expenditure, hormones and appetite profile in the postprandial state. Br. J. Nutr. 2009,
101, 1360�1368. [CrossRef] [PubMed]
97. Taylor, M.A.; Garrow, J.S. Compared with nibbling, neither gorging nor a morning fast affect short-term
energy balance in obese patients in a chamber calorimeter. Int. J. Obes. Relat. Metab. Disord. 2001, 25, 519�528.
[CrossRef] [PubMed]
98. Smeets, A.J.; Westerterp-Plantenga, M.S. Acute effects on metabolism and appetite profile of one meal
difference in the lower range of meal frequency. Br. J. Nutr. 2008, 99, 1316�1321. [CrossRef] [PubMed]
99. Heden, T.D.; LeCheminant, J.D.; Smith, J.D. Influence of weight classification on walking and jogging energy
expenditure prediction in women. Res. Q. Exerc. Sport 2012, 83, 391�399. [CrossRef] [PubMed]
100. Bachman, J.L.; Raynor, H.A. Effects of manipulating eating frequency during a behavioral weight loss
intervention: A pilot randomized controlled trial. Obesity 2012, 20, 985�992. [CrossRef] [PubMed]
101. Perrigue, M.M.; Drewnowski, A.; Wang, C.Y.; Neuhouser, M.L. Higher Eating Frequency Does Not Decrease
Appetite in Healthy Adults. J. Nutr. 2016, 146, 59�64. [CrossRef] [PubMed]
102. Keys, A. Coronary heart disease in seven countries. 1970. Nutrition 1997, 13, 249�253. [CrossRef]
103. Keys, A.; Menotti, A.; Aravanis, C.; Blackburn, H.; Djordevic, B.S.; Buzina, R.; Dontas, A.S.; Fidanza, F.;
Karvonen, M.J.; Kimura, N.; et al. The seven countries study: 2289 deaths in 15 years. Prev. Med. 1984, 13,
141�154. [CrossRef]
104. Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet; a Literature Review.
Nutrients 2015, 7, 9139�9153. [CrossRef] [PubMed]
105. Sofi, F.; Macchi, C.; Abbate, R.; Gensini, G.F.; Casini, A. Mediterranean diet and health status: An updated
meta-analysis and a proposal for a literature-based adherence score. Public Health Nutr. 2014, 17, 2769�2782.
[CrossRef] [PubMed]
106. Mayneris-Perxachs, J.; Sala-Vila, A.; Chisaguano, M.; Castellote, A.I.; Estruch, R.; Covas, M.I.; Fito, M.;
Salas-Salvado, J.; Martinez-Gonzalez, M.A.; Lamuela-Raventos, R.; et al. Effects of 1-year intervention with
a Mediterranean diet on plasma fatty acid composition and metabolic syndrome in a population at high
cardiovascular risk. PLoS ONE 2014, 9, e85202. [CrossRef] [PubMed]
107. Esposito, K.; Maiorino, M.I.; Bellastella, G.; Chiodini, P.; Panagiotakos, D.; Giugliano, D. A journey
into a Mediterranean diet and type 2 diabetes: A systematic review with meta-analyses. BMJ Open
2015, 5, e008222. [CrossRef] [PubMed]
108. Kastorini, C.M.; Milionis, H.J.; Esposito, K.; Giugliano, D.; Goudevenos, J.A.; Panagiotakos, D.B. The effect of
Mediterranean diet on metabolic syndrome and its components: A meta-analysis of 50 studies and 534,906
individuals. J. Am. Coll. Cardiol. 2011, 57, 1299�1313. [CrossRef] [PubMed]
109. Schwingshackl, L.; Missbach, B.; Konig, J.; Hoffmann, G. Adherence to a Mediterranean diet and risk of
diabetes: A systematic review and meta-analysis. Public Health Nutr. 2015, 18, 1292�1299. [CrossRef]
[PubMed]
110. Koloverou, E.; Esposito, K.; Giugliano, D.; Panagiotakos, D. The effect of Mediterranean diet on the
development of type 2 diabetes mellitus: A meta-analysis of 10 prospective studies and 136,846 participants.
Metab. Clin. Exp. 2014, 63, 903�911. [CrossRef] [PubMed]
111. Salas-Salvado, J.; Garcia-Arellano, A.; Estruch, R.; Marquez-Sandoval, F.; Corella, D.; Fiol, M.;
Gomez-Gracia, E.; Vinoles, E.; Aros, F.; Herrera, C.; et al. Components of the Mediterranean-type food
pattern and serum inflammatory markers among patients at high risk for cardiovascular disease. Eur. J.
Clin. Nutr. 2008, 62, 651�659. [CrossRef] [PubMed]
112. Martinez-Gonzalez, M.A.; Garcia-Lopez, M.; Bes-Rastrollo, M.; Toledo, E.; Martinez-Lapiscina, E.H.;
Delgado-Rodriguez, M.; Vazquez, Z.; Benito, S.; Beunza, J.J. Mediterranean diet and the incidence of
cardiovascular disease: A Spanish cohort. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 237�244. [CrossRef]
[PubMed]
113. Fito, M.; Estruch, R.; Salas-Salvado, J.; Martinez-Gonzalez, M.A.; Aros, F.; Vila, J.; Corella, D.; Diaz, O.;
Saez, G.; de la Torre, R.; et al. Effect of the Mediterranean diet on heart failure biomarkers: A randomized
sample from the PREDIMED trial. Eur. J. Heart Fail. 2014, 16, 543�550. [CrossRef] [PubMed]
114. Estruch, R.; Ros, E.; Salas-Salvado, J.; Covas, M.I.; Corella, D.; Aros, F.; Gomez-Gracia, E.; Ruiz-Gutierrez, V.;
Fiol, M.; Lapetra, J.; et al. Primary prevention of cardiovascular disease with a Mediterranean diet. N. Engl.
J. Med. 2013, 368, 1279�1290. [CrossRef] [PubMed]
115. Serra-Majem, L.; Roman, B.; Estruch, R. Scientific evidence of interventions using the Mediterranean diet:
A systematic review. Nutr. Rev. 2006, 64, S27�S47. [CrossRef] [PubMed]
116. Esposito, K.; Kastorini, C.M.; Panagiotakos, D.B.; Giugliano, D. Mediterranean diet and weight loss:
Meta-analysis of randomized controlled trials. Metab. Syndr. Relat. Disord. 2011, 9, 1�12. [CrossRef]
[PubMed]
117. Razquin, C.; Martinez, J.A.; Martinez-Gonzalez, M.A.; Mitjavila, M.T.; Estruch, R.; Marti, A. A 3 years
follow-up of a Mediterranean diet rich in virgin olive oil is associated with high plasma antioxidant capacity
and reduced body weight gain. Eur. J. Clin. Nutr. 2009, 63, 1387�1393. [CrossRef] [PubMed]
118. Bertoli, S.; Spadafranca, A.; Bes-Rastrollo, M.; Martinez-Gonzalez, M.A.; Ponissi, V.; Beggio, V.; Leone, A.;
Battezzati, A. Adherence to the Mediterranean diet is inversely related to binge eating disorder in patients
seeking a weight loss program. Clin. Nutr. 2015, 34, 107�114. [CrossRef] [PubMed]
119. Rios-Hoyo, A.; Cortes, M.J.; Rios-Ontiveros, H.; Meaney, E.; Ceballos, G.; Gutierrez-Salmean, G. Obesity,
Metabolic Syndrome, and Dietary Therapeutical Approaches with a Special Focus on Nutraceuticals
(Polyphenols): A Mini-Review. Int. J. Vitam. Nutr. Res. 2014, 84, 113�123. [CrossRef] [PubMed]
120. Juraschek, S.P.; Guallar, E.; Appel, L.J.; Miller, E.R., 3rd. Effects of vitamin C supplementation on blood
pressure: A meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2012, 95, 1079�1088. [CrossRef]
[PubMed]
121. Michels, A.J.; Frei, B. Myths, artifacts, and fatal flaws: Identifying limitations and opportunities in vitamin C
research. Nutrients 2013, 5, 5161�5192. [CrossRef] [PubMed]
122. Frei, B.; Birlouez-Aragon, I.; Lykkesfeldt, J. Authors� perspective: What is the optimum intake of vitamin C
in humans? Crit. Rev. Food Sci. Nutr. 2012, 52, 815�829. [CrossRef] [PubMed]
123. Mason, S.A.; della Gatta, P.A.; Snow, R.J.; Russell, A.P.; Wadley, G.D. Ascorbic acid supplementation
improves skeletal muscle oxidative stress and insulin sensitivity in people with type 2 diabetes: Findings of
a randomized controlled study. Free Radic. Biol. Med. 2016, 93, 227�238. [CrossRef] [PubMed]
124. Chambial, S.; Dwivedi, S.; Shukla, K.K.; John, P.J.; Sharma, P. Vitamin C in Disease Prevention and Cure:
An Overview. Indian J. Clin. Biochem. 2013, 28, 314�328. [CrossRef] [PubMed]
125. Block, G.; Jensen, C.D.; Dalvi, T.B.; Norkus, E.P.; Hudes, M.; Crawford, P.B.; Holland, N.; Fung, E.B.;
Schumacher, L.; Harmatz, P. Vitamin C treatment reduces elevated C-reactive protein. Free Radic. Biol. Med.
2009, 46, 70�77. [CrossRef] [PubMed]
126. Ashor, A.W.; Siervo, M.; Lara, J.; Oggioni, C.; Afshar, S.; Mathers, J.C. Effect of vitamin C and vitamin E
supplementation on endothelial function: A systematic review and meta-analysis of randomised controlled
trials. Br. J. Nutr. 2015, 113, 1182�1194. [CrossRef] [PubMed]
127. Kim, S.M.; Lim, S.M.; Yoo, J.A.; Woo, M.J.; Cho, K.H. Consumption of high-dose vitamin C (1250 mg
per day) enhances functional and structural properties of serum lipoprotein to improve anti-oxidant,
anti-atherosclerotic, and anti-aging effects via regulation of anti-inflammatory microRNA. Food Funct.
2015, 6, 3604�3612. [CrossRef] [PubMed]
128. Monfared, S.; Larijani, B.; Abdollahi, M. Islet transplantation and antioxidant management: A comprehensive
review. World J. Gastroenterol. 2009, 15, 1153�1161. [CrossRef]
129. German Nutrition Society (DGE). New Reference Values for Vitamin C Intake. Ann. Nutr. Metab. 2015,
67, 13�20.
130. Mamede, A.C.; Tavares, S.D.; Abrantes, A.M.; Trindade, J.; Maia, J.M.; Botelho, M.F. The role of vitamins in
cancer: A review. Nutr. Cancer 2011, 63, 479�494. [CrossRef] [PubMed]
131. Moser, M.A.; Chun, O.K. Vitamin C and Heart Health: A Review Based on Findings from Epidemiologic
Studies. Int. J. Mol. Sci. 2016, 17, 1328. [CrossRef] [PubMed]
132. Vilaplana-Perez, C.; Aunon, D.; Garcia-Flores, L.A.; Gil-Izquierdo, A. Hydroxytyrosol and potential uses in
cardiovascular diseases, cancer, and AIDS. Front. Nutr. 2014, 1, 18. [PubMed]
133. Achmon, Y.; Fishman, A. The antioxidant hydroxytyrosol: Biotechnological production challenges and
opportunities. Appl. Microbiol. Biotechnol. 2015, 99, 1119�1130. [CrossRef] [PubMed]
134. Bulotta, S.; Celano, M.; Lepore, S.M.; Montalcini, T.; Pujia, A.; Russo, D. Beneficial effects of the olive
oil phenolic components oleuropein and hydroxytyrosol: Focus on protection against cardiovascular and
metabolic diseases. J. Transl. Med. 2014, 12, 219. [CrossRef] [PubMed]
135. EFSA NDA Panel (EFSA Panel on Dietetic Products, Nutrition and Allergies). Scientific Opinion on the
substantiation of health claims related to polyphenols in olive and protection of LDL particles from oxidative
damage (ID 1333, 1638, 1639, 1696, 2865), maintenance of normal blood HDL cholesterol concentrations
(ID 1639). EFSA J. 2011, 9, 2033�2058.
136. Scoditti, E.; Nestola, A.; Massaro, M.; Calabriso, N.; Storelli, C.; De Caterina, R.; Carluccio, M.A.
Hydroxytyrosol suppresses MMP-9 and COX-2 activity and expression in activated human monocytes
via PKCalpha and PKCbeta1 inhibition. Atherosclerosis 2014, 232, 17�24. [CrossRef] [PubMed]
137. Giordano, E.; Dangles, O.; Rakotomanomana, N.; Baracchini, S.; Visioli, F. 3-O-Hydroxytyrosol glucuronide
and 4-O-hydroxytyrosol glucuronide reduce endoplasmic reticulum stress in vitro. Food Funct. 2015, 6,
3275�3281. [CrossRef] [PubMed]
138. Granados-Principal, S.; Quiles, J.L.; Ramirez-Tortosa, C.L.; Sanchez-Rovira, P.; Ramirez-Tortosa, M.C.
Hydroxytyrosol: From laboratory investigations to future clinical trials. Nutr. Rev. 2010, 68, 191�206.
[CrossRef] [PubMed]
139. Carluccio, M.A.; Siculella, L.; Ancora, M.A.; Massaro, M.; Scoditti, E.; Storelli, C.; Visioli, F.;
Distante, A.; De Caterina, R. Olive oil and red wine antioxidant polyphenols inhibit endothelial activation:
Antiatherogenic properties of Mediterranean diet phytochemicals. Arterioscler. Thromb. Vasc. Biol. 2003, 23,
622�629. [CrossRef] [PubMed]
140. Visioli, F.; Bernardini, E. Extra virgin olive oil�s polyphenols: Biological activities. Curr. Pharm. Des. 2011, 17,
786�804. [CrossRef] [PubMed]
141. Nabavi, S.F.; Russo, G.L.; Daglia, M.; Nabavi, S.M. Role of quercetin as an alternative for obesity treatment:
You are what you eat! Food Chem. 2015, 179, 305�310. [CrossRef] [PubMed]
142. Vinayagam, R.; Xu, B. Antidiabetic properties of dietary flavonoids: A cellular mechanism review.
Nutr. Metab. 2015, 12, 60. [CrossRef] [PubMed]
143. Shibata, T.; Nakashima, F.; Honda, K.; Lu, Y.J.; Kondo, T.; Ushida, Y.; Aizawa, K.; Suganuma, H.; Oe, S.;
Tanaka, H.; et al. Toll-like receptors as a target of food-derived anti-inflammatory compounds. J. Biol. Chem.
2014, 289, 32757�32772. [CrossRef] [PubMed]
144. Ahn, J.; Lee, H.; Kim, S.; Park, J.; Ha, T. The anti-obesity effect of quercetin is mediated by the AMPK and
MAPK signaling pathways. Biochem. Biophys. Res. Commun. 2008, 373, 545�549. [CrossRef] [PubMed]
145. Fang, X.K.; Gao, J.; Zhu, D.N. Kaempferol and quercetin isolated from Euonymus alatus improve glucose
uptake of 3T3-L1 cells without adipogenesis activity. Life Sci. 2008, 82, 615�622. [CrossRef] [PubMed]
146. Clark, J.L.; Zahradka, P.; Taylor, C.G. Efficacy of flavonoids in the management of high blood pressure.
Nutr. Rev. 2015, 73, 799�822. [CrossRef] [PubMed]
147. D�Andrea, G. Quercetin: A flavonol with multifaceted therapeutic applications? Fitoterapia 2015, 106, 256�271.
[CrossRef] [PubMed]
148. Larson, A.; Witman, M.A.; Guo, Y.; Ives, S.; Richardson, R.S.; Bruno, R.S.; Jalili, T.; Symons, J.D. Acute,
quercetin-induced reductions in blood pressure in hypertensive individuals are not secondary to lower
plasma angiotensin-converting enzyme activity or endothelin-1: Nitric oxide. Nutr. Res. 2012, 32, 557�564.
[CrossRef] [PubMed]
149. Tome-Carneiro, J.; Gonzalvez, M.; Larrosa, M.; Yanez-Gascon, M.J.; Garcia-Almagro, F.J.; Ruiz-Ros, J.A.;
Tomas-Barberan, F.A.; Garcia-Conesa, M.T.; Espin, J.C. Resveratrol in primary and secondary prevention of
cardiovascular disease: A dietary and clinical perspective. Ann. N. Y. Acad. Sci. 2013, 1290, 37�51. [CrossRef]
[PubMed]
150. Leonard, S.S.; Xia, C.; Jiang, B.H.; Stinefelt, B.; Klandorf, H.; Harris, G.K.; Shi, X. Resveratrol scavenges
reactive oxygen species and effects radical-induced cellular responses. Biochem. Biophys. Res. Commun. 2003,
309, 1017�1026. [CrossRef] [PubMed]
151. Ren, Z.; Wang, L.; Cui, J.; Huoc, Z.; Xue, J.; Cui, H.; Mao, Q.; Yang, R. Resveratrol inhibits NF-?B signaling
through suppression of p65 and I?B kinase activities. Die Pharm. 2013, 68, 689�694.
152. Latruffe, N.; Lancon, A.; Frazzi, R.; Aires, V.; Delmas, D.; Michaille, J.J.; Djouadi, F.; Bastin, J.;
Cherkaoui-Malki, M. Exploring new ways of regulation by resveratrol involving miRNAs, with emphasis on
inflammation. Ann. N. Y. Acad. Sci. 2015, 1348, 97�106. [CrossRef] [PubMed]
153. Hausenblas, H.A.; Schoulda, J.A.; Smoliga, J.M. Resveratrol treatment as an adjunct to pharmacological
management in type 2 diabetes mellitus�Systematic review and meta-analysis. Mol. Nutr. Food Res. 2015,
59, 147�159. [CrossRef] [PubMed]
154. Liu, K.; Zhou, R.; Wang, B.; Mi, M.T. Effect of resveratrol on glucose control and insulin sensitivity:
A meta-analysis of 11 randomized controlled trials. Am. J. Clin. Nutr. 2014, 99, 1510�1519. [CrossRef]
[PubMed]
155. Bitterman, J.L.; Chung, J.H. Metabolic effects of resveratrol: Addressing the controversies. Cell. Mol. Life Sci.
2015, 72, 1473�1488. [CrossRef] [PubMed]
156. Han, S.; Park, J.S.; Lee, S.; Jeong, A.L.; Oh, K.S.; Ka, H.I.; Choi, H.J.; Son, W.C.; Lee, W.Y.; Oh, S.J.; et al.
CTRP1 protects against diet-induced hyperglycemia by enhancing glycolysis and fatty acid oxidation.
J. Nutr. Biochem. 2016, 27, 43�52. [CrossRef] [PubMed]
157. Gambini, J.; Ingles, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.;
Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo
Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxid. Med.
Cell. Longev. 2015, 2015, 837042. [CrossRef] [PubMed]
158. Yang, C.S.; Suh, N. Cancer prevention by different forms of tocopherols. Top. Curr. Chem. 2013, 329, 21�33.
[PubMed]
159. Jiang, Q. Natural forms of vitamin E: Metabolism, antioxidant, and anti-inflammatory activities and their
role in disease prevention and therapy. Free Radic. Biol. Med. 2014, 72, 76�90. [CrossRef] [PubMed]
160. Witting, P.K.; Upston, J.M.; Stocker, R. The molecular action of alpha-tocopherol in lipoprotein lipid
peroxidation. Pro- and antioxidant activity of vitamin E in complex heterogeneous lipid emulsions.
In Fat-Soluble Vitamins; Quinn, P.J., Kagan, V.E., Eds.; Springer: New York, NY, USA; pp. 345�390.
161. Saboori, S.; Shab-Bidar, S.; Speakman, J.R.; Yousefi Rad, E.; Djafarian, K. Effect of vitamin E supplementation
on serum C-reactive protein level: A meta-analysis of randomized controlled trials. Eur. J. Clin. Nutr. 2015,
69, 867�873. [CrossRef] [PubMed]
162. Azzi, A.; Meydani, S.N.; Meydani, M.; Zingg, J.M. The rise, the fall and the renaissance of vitamin E.
Arch. Biochem. Biophys. 2016, 595, 100�108. [CrossRef] [PubMed]
163. Raederstorff, D.; Wyss, A.; Calder, P.C.; Weber, P.; Eggersdorfer, M. Vitamin E function and requirements in
relation to PUFA. Br. J. Nutr. 2015, 114, 1113�1122. [CrossRef] [PubMed]
164. Loffredo, L.; Perri, L.; Di Castelnuovo, A.; Iacoviello, L.; De Gaetano, G.; Violi, F. Supplementation
with vitamin E alone is associated with reduced myocardial infarction: A meta-analysis. Nutr. Metab.
Cardiovasc. Dis. 2015, 25, 354�363. [CrossRef] [PubMed]
165. Giampieri, F.; Tulipani, S.; Alvarez-Suarez, J.M.; Quiles, J.L.; Mezzetti, B.; Battino, M. The strawberry:
Composition, nutritional quality, and impact on human health. Nutrition 2012, 28, 9�19. [CrossRef] [PubMed]
166. Amiot, M.J.; Riva, C.; Vinet, A. Effects of dietary polyphenols on metabolic syndrome features in humans:
A systematic review. Obes. Rev. 2016, 17, 573�586. [CrossRef] [PubMed]
167. Smeriglio, A.; Barreca, D.; Bellocco, E.; Trombetta, D. Chemistry, Pharmacology and Health Benefits of
Anthocyanins. Phytother. Res. 2016, 30, 1265�1286. [CrossRef] [PubMed]
168. Lila, M.A. Anthocyanins and Human Health: An In Vitro Investigative Approach. J. Biomed. Biotechnol. 2004,
2004, 306�313. [CrossRef] [PubMed]
169. Stull, A.J.; Cash, K.C.; Johnson, W.D.; Champagne, C.M.; Cefalu, W.T. Bioactives in blueberries improve
insulin sensitivity in obese, insulin-resistant men and women. J. Nutr. 2010, 140, 1764�1768. [CrossRef]
[PubMed]
170. Zhu, Y.; Xia, M.; Yang, Y.; Liu, F.; Li, Z.; Hao, Y.; Mi, M.; Jin, T.; Ling, W. Purified anthocyanin supplementation
improves endothelial function via NO-cGMP activation in hypercholesterolemic individuals. Clin. Chem.
2011, 57, 1524�1533. [CrossRef] [PubMed]
171. Qin, Y.; Xia, M.; Ma, J.; Hao, Y.; Liu, J.; Mou, H.; Cao, L.; Ling, W. Anthocyanin supplementation improves
serum LDL- and HDL-cholesterol concentrations associated with the inhibition of cholesteryl ester transfer
protein in dyslipidemic subjects. Am. J. Clin. Nutr. 2009, 90, 485�492. [CrossRef] [PubMed]
172. Zhu, Y.; Ling, W.; Guo, H.; Song, F.; Ye, Q.; Zou, T.; Li, D.; Zhang, Y.; Li, G.; Xiao, Y.; et al. Anti-inflammatory
effect of purified dietary anthocyanin in adults with hypercholesterolemia: A randomized controlled trial.
Nutr. Metab. Cardiovasc. Dis. 2013, 23, 843�849. [CrossRef] [PubMed]
173. Zhu, Y.; Huang, X.; Zhang, Y.; Wang, Y.; Liu, Y.; Sun, R.; Xia, M. Anthocyanin supplementation
improves HDL-associated paraoxonase 1 activity and enhances cholesterol efflux capacity in subjects
with hypercholesterolemia. J. Clin. Endocrinol. Metab. 2014, 99, 561�569. [CrossRef] [PubMed]
174. Karlsen, A.; Retterstol, L.; Laake, P.; Paur, I.; Bohn, S.K.; Sandvik, L.; Blomhoff, R. Anthocyanins inhibit
nuclear factor-kappaB activation in monocytes and reduce plasma concentrations of pro-inflammatory
mediators in healthy adults. J. Nutr. 2007, 137, 1951�1954. [PubMed]
175. Keske, M.A.; Ng, H.L.; Premilovac, D.; Rattigan, S.; Kim, J.A.; Munir, K.; Yang, P.; Quon, M.J. Vascular and
metabolic actions of the green tea polyphenol epigallocatechin gallate. Curr. Med. Chem. 2015, 22, 59�69.
[CrossRef] [PubMed]
176. Johnson, R.; Bryant, S.; Huntley, A.L. Green tea and green tea catechin extracts: An overview of the clinical
evidence. Maturitas 2012, 73, 280�287. [CrossRef] [PubMed]
177. Huang, J.; Wang, Y.; Xie, Z.; Zhou, Y.; Zhang, Y.; Wan, X. The anti-obesity effects of green tea in human
intervention and basic molecular studies. Eur. J. Clin. Nutr. 2014, 68, 1075�1087. [CrossRef] [PubMed]
178. Hursel, R.; Westerterp-Plantenga, M.S. Catechin- and caffeine-rich teas for control of body weight in humans.
Am. J. Clin. Nutr. 2013, 98, 1682S�1693S. [CrossRef] [PubMed]
179. Gutierrez-Salmean, G.; Ortiz-Vilchis, P.; Vacaseydel, C.M.; Rubio-Gayosso, I.; Meaney, E.; Villarreal, F.;
Ramirez-Sanchez, I.; Ceballos, G. Acute effects of an oral supplement of (?)-epicatechin on postprandial fat
and carbohydrate metabolism in normal and overweight subjects. Food Funct. 2014, 5, 521�527. [CrossRef]
[PubMed]
180. Khalesi, S.; Sun, J.; Buys, N.; Jamshidi, A.; Nikbakht-Nasrabadi, E.; Khosravi-Boroujeni, H. Green tea
catechins and blood pressure: A systematic review and meta-analysis of randomised controlled trials.
Eur. J. Nutr. 2014, 53, 1299�1311. [CrossRef] [PubMed]

Close Accordion
Metabolic Syndrome And Chiropractic

Metabolic Syndrome And Chiropractic

Metabolic Syndrome:

Key indexing terms:

  • Metabolic syndrome X
  • Insulin resistance
  • Hyperglycemia
  • Inflammation
  • Weight loss

Abstract
Objective: This article presents an overview of metabolic syndrome (MetS), which is a collection of risk factors that can lead to diabetes, stroke, and heart disease. The purposes of this article are to describe the current literature on the etiology and pathophysiology of insulin resistance as it relates to MetS and to suggest strategies for dietary and supplemental management in chiropractic practice.

Methods: The literature was searched in PubMed, Google Scholar, and the Web site of the American Heart Association, from the earliest date possible to May 2014. Review articles were identified that outlined pathophysiology of MetS and type 2 diabetes mellitus (T2DM) and relationships among diet, supplements, and glycemic regulation, MetS, T2DM, and musculoskeletal pain.

Results: Metabolic syndrome has been linked to increased risk of developing T2DM and cardiovascular disease and increased risk of stroke and myocardial infarction. Insulin resistance is linked to musculoskeletal complaints both through chronic inflammation and the effects of advanced glycosylation end products. Although diabetes and cardiovascular disease are the most well-known diseases that can result from MetS, an emerging body of evidence demonstrates that common musculoskeletal pain syndromes can be caused by MetS.

Conclusions: This article provides an overview of lifestyle management of MetS that can be undertaken by doctors of chiropractic by means of dietary modification and nutritional support to promote blood sugar regulation.

Introduction: Metabolic Syndrome

Metabolic syndrome (MetS) has been described as a cluster of physical examination and laboratory findings�that directly increases the risk of degenerative metabolic disease expression. Excess visceral adipose tissue, insulin resistance, dyslipidemia, and hypertension are conditions that significantly contribute to the syndrome. These conditions are united by a pathophysiological basis in low-grade chronic inflammation and increase an individual’s risk of cardiovascular disease, type 2 diabetes mellitus (T2DM), and all-cause mortality.1

The National Health and Nutrition Examination Survey (NHANES) 2003-2006 estimated that approximately 34% of United States adults aged 20 years and more had MetS.2 The same NHANES data found that 53% had abdominal adiposity, a condition that is closely linked to visceral adipose stores. Excess visceral adiposity generates increased systemic levels of pro-inflammatory mediator molecules. Chronic, low- grade inflammation has been well documented as an associated and potentially inciting factor for the development of insulin resistance and T2DM.1

NHANES 2003-2006 data showed that 39% of subjects met criteria for insulin resistance. Insulin resistance is a component of MetS that significantly contributes to the expression of chronic, low-grade inflammation and predicts T2DM expression. T2DM costs the United States in excess of $174 billion in 2007. 3 It is estimated that 1 in 4 adults will have T2DM by the year 2050.3 Currently, more than one third of US adults (34.9%) are obese, 4 and, in 2008, the annual medical cost of obesity was $147 billion.4,5 This clearly represents a health care concern.

The pervasiveness of MetS dictates that doctors of chiropractic will see a growing proportion of patients who fit the syndrome criteria.6 Chiropractic is most commonly used for musculoskeletal complaints believed to be mechanical in nature;6 however, an emerging body of evidence identifies MetS as a biochemical promoter of musculoskeletal complaints such as neck pain, shoulder pain, patella tendinopathy, and widespread musculoskeletal pain. 7�13 As an example, the cross-linking of collagen fibers can be caused by increased advanced glycation end-product (AGE) formation as seen in insulin resistance.14 Increased collagen cross-linking is observed in both osteoarthritis and degenerative disc disease, 15 and reduced mobility in elderly patients with T2DM has also been attributed to AGE-induced collagen cross-linking. 16,17

A diagnosis of MetS is made from a patient having 3 of the 5 findings presented in Table 1. Fasting hyperglycemia is termed impaired fasting glucose and indicates insulin resistance. 18,19 An elevated hemoglobin A1c (HbA1c) level measures long-term blood glucose�regulation and is diagnostic for T2DM when elevated in the presence of impaired fasting glucose. 3,18

metabolic table 1

The emerging evidence demonstrates that we cannot view musculoskeletal pain as only coming from conditions that are purely mechanical in nature. Doctors of chiropractic must demonstrate prowess in identification and management of MetS and an understanding of insulin resistance as its main pathophysiological feature. The purposes of this article are to describe the current literature on the etiology and pathophysiology of insulin resistance as it relates to MetS and to suggest strategies for dietary and supplemental management in chiropractic practice.

Methods

metabolic method arrowsPubMed was searched from the earliest possible date to May 2014 to identify review articles that outlined the pathophysiology of MetS and T2DM. This led to further search refinements to identify inflammatory mechanisms that occur in the pancreas, adipose tissue, skeletal muscle, and hypothalamus. Searches were also refined to identify relationships among diet, supplements, and glycemic regulation. Both animal and human studies were reviewed. The selection of specific supplements was based on those that were most commonly used in the clinical setting, namely, gymnema sylvestre, vanadium, chromium and ?-lipoic acid.

Discussion

Insulin Resistance Overview

metabolic insulin resistance 1Under normal conditions, skeletal muscle, hepatic, and adipose tissues require the action of insulin for cellular glucose entry. Insulin resistance represents an inability of insulin to signal glucose passage into insulin-dependent cells. Although a genetic predisposition can exist, the�etiology of insulin resistance has been linked to chronic low-grade inflammation.1 Combined with insulin resistance-induced hyperglycemia, chronic low-grade inflammation also sustains MetS pathophysiology.1

Two thirds of postprandial blood glucose metabolism occurs within skeletal muscle via an insulin-dependent mechanism.18,19 Insulin binding to its receptor triggers glucose entry and subsequently inhibits lipolysis within the target tissue.21,22 Glucose enters skeletal muscles cells by way of a glucose transporter designated Glut4. 18 Owing to genetic variability, insulin-mediated glucose uptake can vary more than 6-fold among non-diabetic individuals. 23

Prolonged insulin resistance leads to structural changes within skeletal muscle such as decreased Glut4 transporter number, intramyocellular fat accu- mulation, and a reduction in mitochondrial con- tent.19,24 These events are thought to impact energy generation and functioning of affected skeletal mus- cle.24 Insulin-resistant skeletal muscle is less able to suppress lipolysis in response to insulin binding.25 Subsequently, saturated free fatty acids accumulate and generate oxidative stress. 22 The same phenomenon within adipose tissue generates a rapid adipose cell expansion and tissue hypoxia.26 Both these processes increase inflammatory pathway activation and the generation of proinflammatory cytokines (PICs).27

Multiple inflammatory mediators are associated with the promotion of skeletal muscle insulin resistance. The PICs tumor necrosis factor ? (TNF-?), interleukin 1 (IL- 1), and IL-6 have received much attention because of their direct inhibition of insulin signaling.28�30 Since cytokine testing is not performed clinically, elevated levels of high- sensitivity C-reactive protein (hsCRP) best represent the low-grade systemic inflammation that characterizes insulin resistance.31,32

Insulin resistance�induced hyperglycemia can lead to irreversible changes in protein structure, termed glycation, and the formation of AGEs. Cells such as those of the vascular endothelium are most vulnerable to hyperglycemia due to utilization of an insulin-independent Glut1 transporter. 33 This makes AGE generation responsible for most diabetic complications, 15,33,34 including collagen cross-linking.15

If unchanged, prolonged insulin resistance can lead to T2DM expression. The relationship between chronic low-grade inflammation and T2DM has been well characterized. 35 Research has demonstrated that patients with T2DM also have chronic inflammation within the pancreas, termed insulitis, and it worsens hyperglycemia due to the progressive loss of insulin- producing ? cells.36�39

Visceral Adiposity And Insulin Resistance

metabolic Visceral Adiposity Insulin resistanceCaloric excess and a sedentary lifestyle contribute to the accumulation of subcutaneous and visceral adipose tissue. Adipose tissue was once thought of as a metabolically inert passive energy depot. A large body of evidence now demonstrates that excess visceral adipose tissue acts as a driver of chronic low-grade inflammation and insulin resistance.27,34

It has been documented that immune cells infiltrate rapidly expanding visceral adipose tissue. 26,40 Infil- trated macrophages become activated and release PICs that ultimately cause a phenotypic shift in resident macrophage phenotype to a classic inflammatory M1 profile.27 This vicious cycle creates a chronic inflam- matory response within adipose tissue and decreases the production of adipose-derived anti-inflammatory cytokines.43 As an example, adiponectin is an adipose- derived anti-inflammatory cytokine. Macrophage- invaded adipose tissue produces less adiponectin, and this has been correlated with increasing insulin resistance. 26

Hypothalamic Inflammation And Insulin Resistance

metabolic Hypothalamic Inflammation And Insulin ResistanceEating behavior in the obese and overweight has been popularly attributed to a lack of will power or genetics. However, recent research has demonstrated a link between hypothalamic inflammation and increased body weight.41,41

Centers that govern energy balance and glucose homeostasis are located within the hypothalamus. Recent studies demonstrate that inflammation in the hypothalamus coincides with metabolic inflammation and an increase in appetite.43 These hypothalamic centers simultaneously become resistant to anorexigenic stimuli, leading to altered energy intake. It has been suggested that this provides a neuropathological basis for MetS and drives a progressive increase in body weight. 41

Central metabolic inflammation pathologically activates hypothalamic immune cells and disrupts central insulin and leptin signaling.41 Peripherally, this has been associated with dysregulated glucose homeostasis that also impairs pancreatic ? cell functioning.41,44 Hypothalamic inflammation contributes to hypertension through similar mechanisms, and it is thought that central inflammation parallels chronic low-grade systemic inflammation and insulin resistance.41�44

Clinical Correlates Diet-Induced Inflammation & Insulin Resistance

Fatty foodsFeeding generally leads to a short-term increase in both oxidative stress and inflammation. 41 Total�calories consumed, glycemic index, and fatty acid profile of a meal all influence the degree of postprandial inflammation. It is estimated that the average American consumes approximately 20% of calories from refined sugar, 20% from refined grains and flour, 15% to 20% from excessively fatty meat products, and 20% from refined seed/legume oils.45 This pattern of eating contains a macronutrient composition and glycemic index that promote hyperglycemia, hyperlipemia, and an acute postprandial inflammatory response. 46 Collectively referred to as postprandial dysmetabolism, this pro-inflammatory response can sustain levels of chronic low-grade inflammation that leads to excess body fat, coronary heart disease (CHD), insulin resistance, and T2DM.28,29,47

Recent evidence suggests that several MetS criteria may not sufficiently identify all individuals with postprandial dysmetabolism. 48,49 A 2-hour oral glucose tolerance test (2-h OGTT) result greater than 200 mg/dL can be used clinically to diagnose T2DM. Although MetS includes a fasting blood glucose level less than 100 mg/dL, population studies have shown that a fasting glucose as low as 90 mg/dL can be associated with an 2-h OGTT level greater than 200 mg/dL.49 Further, a recent large cohort study indicated that an increased 2-h OGTT was independently predictive of cardiovascular and all-cause mortality in a nondiabetic population. 48 Mounting evidence indicates that post- prandial glucose levels are better correlated with MetS and predicting future cardiovascular events than fasting blood glucose alone.41,48

Fasting triglyceride levels generally correlate with postprandial levels, and a fasting triglyceride level greater than 150 mg/dL reflects MetS and insulin resistance. Contrastingly, epidemiologic data indicate that a fasting triglyceride level greater than 100 mg/dL influences CHD risk via postprandial dysmetabolism. 48 The acute postprandial inflammatory response that contributes to CHD risk includes an increase in PICs, free radicals, and hsCRP.48,49 These levels are not measured clinically but, monitoring fasting glucose, 2-hour postprandial glucose and fasting triglycerides can be used as correlates of postprandial dysmetabolic and low-grade systemic inflammation.

MetS And Disease Expression

metabolic diabetes related wordsDiagnosis of MetS has been linked to an increased risk of developing T2DM and cardiovascular disease over the following 5 to 10 years. 1 It further increases a patient’s risk of stroke, myocardial infarction, and death from any of the aforementioned conditions.1

Facchini et al47 followed 208 apparently healthy, non-obese subjects for 4 to 11 years while monitoring the incidence of clinical events such as hypertension, stroke, CHD, cancer, and T2DM. Approximately one fifth of participants experienced clinical events, and all of these subjects were either classified as intermediately or severely insulin resistant. It is important to note that all of these clinical events have a pathological basis in chronic low-grade inflammation,50 and no events were experienced in the insulin-sensitive groupings. 47

Insulin resistance is linked to musculoskeletal com- plaints both through chronic inflammation and the effects of AGEs. Advanced glycation end-products have been shown to extensively accumulate in osteoarthritic cartilage and treatment of human chondrocytes with AGEs increased their catabolic activity. 51 Advanced glycation end-products increase collagen stiffness via cross-linking and likely contribute to reduced joint mobility seen in elderly patients with T2DM.52 Com- pared to non-diabetics, type II diabetic patients are known to have altered proteoglycan metabolism in their intervertebral discs. This altered metabolism may pro- mote weakening of the annular fibers and subsequently, disc herniation.53 The presence of T2DM increases a person’s risk of expressing disc herniation in both the cervical and lumbar spines.17,54 Patients with T2DM are also more likely to develop lumbar stenosis compared with non-diabetics, and this has been documented as a plausible relationship between MetS risk factors and physician-diagnosed lumbar disc herniation. 55�57

There are no specific symptoms that denote early skeletal muscle structural changes. Fatty infiltration and decreased muscle mitochondria content are observed within age-related sarcopenia 58 ; however, it is still being argued whether fatty infiltration is a risk factor for low back pain. 59,60

Clinical management of MetS should be geared toward improving insulin sensitivity and reducing chronic low-grade inflammation. 1 Regular exercise without weight loss is associated with reduced insulin resistance, and at least 30 minutes of aerobic activity and resistance training is recommended daily. 61,62 Although frequently considered preventative, exercise, dietary, and weight loss interventions should be considered alongside pharmacological management in those with MetS. 1

Data regarding the exact amount of weight loss needed to improve chronic inflammation are inconclusive. In overweight individuals without diagnosed MetS, a very-low-carbohydrate diet (b 10% calories from carbohydrate) has significantly reduced plasma inflammatory markers (TNF-?, hsCRP, and IL-6) with�as little as 6% reduction in body weight.63,64 Individuals who meet MetS criteria may require 10% to 20% body weight loss to reduce inflammatory markers. 65 Interestingly, the Mediterranean Diet has been shown to reduce markers of systemic inflammation independent of weight loss65 and was recommended in the American College of Cardiology and American Heart Association Adult Treatment Panel 4 guidelines.66

A growing body of research has examined the effects of the Spanish ketogenic Mediterranean diet, including olive oil, green vegetables and salads, fish as the primary protein, and moderate red wine consumption. In a sample of 22 patients, adoption of the Spanish ketogenic Mediterranean diet with 9 g of supplemental salmon oil on days when fish was not consumed has led to complete resolution of MetS.67 Significant reductions in markers of chronic systemic inflammation were seen in 31 patients following this diet for 12 weeks.68

A Paleolithic diet based on lean meat, fish, fruits, vegetables, root vegetables, eggs, and nuts has been described as more satiating per calorie than a diabetes diet in patients with T2DM.69 In a randomized crossover study, a Paleolithic diet resulted in lower mean HbA1c values, triglycerides, diastolic blood pressure, waist circumference, improved glucose tolerance, and higher high-density lipoprotein (HDL) values compared to a diabetes diet.70 Within the context of these changes, a referral for medication management may be advisable.

Irrespective of name, a low-glycemic diet that focuses on vegetables, fruits, lean meats, omega-3 fish, nuts, and tubers can be considered anti-inflammatory and has been shown to ameliorate insulin resistance. 49,71�73 Inflammatory markers and insulin resistance further improve when weight loss coincides with adherence to an anti-inflammatory diet.70 A growing body of evidence suggests that specific supplemental nutrients also reduce insulin resistance and improve chronic low-grade inflammation.

Key Nutrients That Promote Insulin Sensitivity

metabolic nutrientsResearch has identified nutrients that play key roles in promoting proper insulin sensitivity, including vitamin D, magnesium, omega-3 (n-3) fatty acids, curcumin, gymnema, vanadium, chromium, and ?-lipoic acid. It is possible to get adequate vitamin D from sun exposure and adequate amounts of magnesium and omega-3 fatty acids from food. Contrastingly, the therapeutic levels of chromium and ?-lipoic acid that affect insulin sensitivity and reduce�insulin resistance cannot be obtained in food and must be supplemented.

Vitamin D, Magnesium, Omega-3 Fatty Acids, & Curcumin

metabolic Vitamin D, Magnesium, Omega-3 Fatty Acids, CurcuminVitamin D, magnesium, and n-3 fatty acids have multiple functions, and generalized inflammation reduction is a common mechanism of action.74�80 Their supplemental use should be considered in the context of low-grade inflammation reduction and health promotion, rather than as a specific treatment for MetS or T2DM.

Evidence pertaining to the precise role of vitamin D in MetS and insulin resistance is inconclusive. Increas- ing dietary and supplemental vitamin D intake in young men and women may lower the risk of MetS and T2DM development,81 and a low serum vitamin D level has been associated with insulin resistance and T2DM expression. 82 Supplementation to improve low serum vitamin D (reference range, 32-100 ng/mL) is effective, but its impact on improving central glycemia and insulin sensitivity is conflicting. 83 Treating insulin resistance and MetS with vitamin D as a monotherapy appears to be unsuccessful. 82,83 Achieving normal vitamin D blood levels through adequate sun exposure and/or supplementation is advised for general health. 84�86

The average American diet commonly contains a low magnesium intake.80 Recent studies suggest that supple- mental magnesium can improve insulin sensitivity. 81,82 Taking 365 mg/d may be effective in reducing fasting glucose and raising HDL cholesterol in T2DM,83 as well as normomagnesemic, overweight, nondiabetics. 84

Diets high in the omega-6 fat linoleic acid have been associated with insulin resistance85 and higher levels of serum pro-inflammatory mediator markers including IL-6, IL-1?, TNF-?, and hsCRP.87 Supplementation to increase dietary omega-3 fatty acids at the expense of omega-6 fatty acids has been shown to improve insulin sensitivity. 88�90 Six months of omega-3 supplementation at 3 g/d with meals has been shown to reduce MetS markers including fasting triglycerides, HDL cholesterol, and an increase in anti-inflammatory adiponectin. 91

Curcumin is responsible for the yellow pigmentation of the spice turmeric. Its biological effects can be characterized as antidiabetic and antiobesity via down- regulating TNF-?, suppressing nuclear factor ?B activation, adipocytokine expression, and leptin level modulation,. 92�95 Curcumin has been reported to activate peroxisome proliferator-activated receptor-?, the nuclear target of the thiazolidinedione class of antidiabetic drugs,93 and it also protects hepatic and pancreatic cells. 92,93 Numerous studies have reported�weight loss, hsCRP reduction, and improved insulin sensitivity after curcumin supplementation.92�95

There is no established upper limit for curcumin, and doses of up to 12 g/d are safe and tolerable in humans. 96 A randomized, double-blinded, placebo- controlled trial (N = 240) showed a reduced progression of prediabetes to T2DM after 9 months of 1500 mg/d curcumin supplementation.97

Curcumin, 98 vitamin D, 84 magnesium, 91 and omega-3 fatty acids80 are advocated as daily supplements to promote general health. A growing body of evidence supports the views of Gymnema sylvestre, vanadium, chromium, and ?-lipoic acid should as therapeutic supplements to assist in glucose homeostasis.

G Sylvestre

metabolic Gymnema sylvestre medicinal herbGymnemic acids are the active component of the G sylvestre plant leaves. Gymnemic acids are the active component of the G sylvestre plant leaves. Studies evaluating G sylvestre’s effects on diabetes in humans have generally been of poor methodological quality. Experimental animal studies have found that gymnemic acids may decrease glucose uptake in the small intestine, inhibit gluconeogenesis, and reduce hepatic and skeletal muscle insulin resistance.99 Other animal studies suggest that gymnemic acids may have comparable efficacy in reducing blood sugar levels to the first-generation sulfonylurea, tolbutamide.100

Evidence from open-label trials suggests its use as a supplement to oral antidiabetic hypoglycemic agents. 96 One quarter of patients were able to discontinue their drug and maintain normal glucose levels on an ethanolic gymnema extract alone. Although the evidence to date suggests its use in humans and animals is safe and well tolerated, higher quality human studies are warranted.

Vanadyl Sulfate

metabolic Vanadyl SulfateVanadyl sulfate has been reported to prolong the events of insulin signaling and may actually improve insulin sensitivity.101 Limited data suggest that it inhibits gluconeogenesis, possibly ameliorating hepatic insulin resistance. 100,101 Uncontrolled clinical trials have reported improvements in insulin sensitivity using 50 to 300 mg daily for periods ranging from 3 to 6 weeks. 101�103 Contrastingly, a recent randomized, double-blind, placebo-controlled trial found that 50 mg of vanadyl sulfate twice daily for 4 weeks had no effect in individuals with impaired glucose tolerance. 104 Limited clinical and experimental data exist supporting the use of vanadyl sulfate to improve insulin resistance,�and further research is warranted regarding its safety and efficacy.

Chromium

metabolic ChromiumDiets high in refined sugar and flour are deficient in chromium (Cr) and lead to an increased urinary excretion of chromium. 105,106 The progression of MetS is not likely caused by a chromium deficiency, 107 and dosages that benefit glycemic regulation are not achievable through food. 106,108,109

A recent randomize, double-blind trial demonstrated that 1000 ?g Cr per day for 8 months improved insulin sensitivity by 10% in subjects with T2DM.110 Cefalu et al110 further suggested that these improvements might be more applicable to patients with a greater degree of insulin resistance, impaired fasting plasma glucose, and higher HbA1c values. Chromium’s mechanism of action for improving insulin sensitivity is through increased Glut4 translocation via prolonging insulin receptor signaling.109 Chromium has been well tolerated at 1000 ?g/d,105 and animal models using significantly more than 1000 ? Cr per day were not associated with toxicological consequences.109

?-Lipoic Acid

metabolic alpha-lipoic-acidHumans derive ?-lipoic acid through dietary means and from endogenous synthesis. 111 The foods richest in ?-lipoic acid are animal tissues with extensive metabolic activity such as animal heart, liver, and kidney, which are not consumed in large amounts in the typical American diet. 111 Supplemental amounts of ?-lipoic acid used in the treatment of T2DM (300-600 mg) are likely to be as much as 1000 times greater than the amounts that could be obtained from the diet.112

Lipoic acid synthase (LASY) appears to be the key enzyme involved in the generation of endogenous lipoic acid, and obese mice with diabetes have reduced LASY expression when compared with age-and sex- matched controls.111 In vitro studies to identify potential inhibitors of lipoic acid synthesis suggest a role for diet-induced hyperglycemia and the PIC TNF- ? in the down-regulation of LASY.113 The inflammatory basis of insulin resistance may therefore drive lowered levels of endogenous lipoic acid via reducing the activity of LASY.

?-Lipoic acid has been found to act as insulin mimetic via stimulating Glut4-mediated glucose trans- port in muscle cells. 110,114?-Lipoic acid is a lipophilic free radical scavenger and may affect glucose homeostasis through protecting the insulin receptor from damage114 and indirectly via decreasing nuclear factor ?B�mediated TNF-? and IL-1 production. 110 In�postmenopausal women with MetS (presence of at least 3 ATPIII clinical criteria) 4 g/d of a combined inositol and ?-lipoic acid supplement for 6 months significantly improved OGTT scores by 20% in two thirds of the subjects. 114 A recent randomized double-blinded placebo-controlled study showed that 300 mg/d ?- lipoic acid for 90 days significantly decreased HbA1c values in subjects with T2DM.115

Side effects to ?-lipoic acid supplementation as high as 1800 mg/d have largely been limited to nausea. 116 It may be best to take supplemental ?-lipoic acid on an empty stomach (1 hour before or 2 hours after eating) because food intake reportedly reduces its bioavailability.117 Clinicians should be aware that ?-lipoic acid supplementation might increase the risk of hypoglycemia in diabetic patients using insulin or oral antidiabetic agents.117

Limitations

metabolic limitations signThis is a narrative overview of the topic of MetS. A systematic review was not performed; therefore, there may be relevant information missing from this review. The contents of this overview focuses on the opinions of the authors, and therefore, others may disagree with our opinions or approaches to management. This overview is limited by the studies that have been published. To date, no studies have been published that identify the effectiveness of a combination of a dietary intervention, such as the Spanish ketogenic diet, and nutritional supplementation on the expression of the MetS. Similarly, this approach has not been studied in patients with musculoskeletal pain who also have the MetS. Consequently, the information presented in this article is speculative. Longitudinal studies are needed before any specific recommendations can be made for patients with musculoskeletal that may be influenced by the MetS.

Conclusion: Metabolic Syndrome

This overview suggests that MetS and type 2 diabetes are complex conditions, and their prevalence is expected to increase substantially in the coming years. Thus, it is important to identify if the MetS may be present in patients who are nonresponsive to manual care and to help predict who may not respond adequately.

We suggest that diet and exercise are essential to managing these conditions, which can be supported with key nutrients, such as vitamin D, magnesium, and�omega-3 fatty acids. We also suggest that curcumin, G sylvestre, vanadyl sulfate chromium, and ?-lipoic acid could be viewed as specific nutrients that may be taken during the process of restoring appropriate insulin sensitivity and signaling.

Chiropractic Care

 

David R. Seaman DC, MS,?, Adam D. Palombo DC

Professor, Department of Clinical Sciences, National University of Health Sciences, Pinellas Park, FL Private Chiropractic Practice, Newburyport, MA

Funding Sources and Conflicts of Interest

No funding sources were reported for this study. David Seaman is a paid consultant for Anabolic Laboratories, a manufacturer of nutritional products for health care professionals. Adam Palombo was sponsored and remunerated by Anabolic laboratories to speak at chiropractic conventions/meetings.

Blank
References:

1. Kaur J. A comprehensive review on metabolic syndrome.<br />
Cardiol Res Pract 2014:943162, dx.doi.org/10.1155/<br />
2014/943162.<br />
2. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic<br />
syndrome among US adults. Findings from the Third National<br />
Health and Nutrition Examination Survey. J Am Med Assoc<br />
2006;287:356�9.<br />
3. Boyle JP, Thompson TJ, Gregg EW, Barker LE, Williamson<br />
DF. Projection of the year 2050 burden of diabetes in the US<br />
adult population: dynamic modeling of incidence, mortality,<br />
and prediabetes prevalence. Popul Health Metr 2010;8:29,<br />
dx.doi.org/10.1186/1478-7954-8-29.<br />
4. [Internet]Centers for Disease Control and Prevention.<br />
Adult Obesity Facts. Atlanta: CDC; 2014. [Available from<br />www.cdc.gov/obesity/data/adult.html%5D.<br />
5. Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of<br />
childhood and adult obesity in the United States, 2011�2012.<br />
JAMA 2014;311(8):806�14.<br />
6. Riksman JS, Williamson OD, Walker BF. Delineating<br />
inflammatory and mechanical sub-types of low back pain: a<br />
pilot survey of fifty low back pain patients in a chiropractic<br />
setting. Chiropr Man Therap 2011;19(1):5, dx.doi.org/<br />
10.1186/2045-709X-19-5.<br />
7. Dobretsov M, Ghaleb AH, Romanovsky D, Pablo CS, Stimers<br />
JR. Impaired insulin signaling as a potential trigger of<br />
pain in diabetes and prediabetes. Int Anesthesiol Clin<br />
2007;45(2):95�105.<br />
8. Mantyselka P, Miettola J, Niskanen L, Kumpusalo E. Glucose<br />
regulation and chronic pain at multiple sites. Rheumatology<br />
2008;47(8):1235�8.<br />
9. M�ntyselk� P, Miettola J, Niskanen L, Kumpusalo E.<br />
Persistent pain at multiple sites�connection to glucose<br />
derangement. Diabetes Res Clin Pract 2009;84(2):e30�2.<br />
10. Mantyselka P, Kautianen H, Vanhala M. Prevalence of neck<br />
pain in subjects with metabolic syndrome�a cross-sectional<br />
population-based study. BMC Musculoskelet Disord 2010;11:<br />
171, dx.doi.org/10.1186/1471-2474-11-171.<br />
11. Rechardt M, Shiri R, Karppinen J, Jula A, Heli�vaara M,<br />
Viikari-Juntura E. Lifestyle and metabolic factors in relation<br />
to shoulder pain and rotator cuff tendinitis: a population-based<br />
study. BMC Musculoskelet Disord 2010;11:165.<br />
12. Gaida JE, Alfredson L, Kiss ZS, Wilson AM, Alfredson H,<br />
Cook JL. Dyslipidemia in Achilles tendinopathy is<br />
characteristic of insulin resistance. Med Sci Sports Exerc<br />
2009;41:1194�7.<br />
13. Malliaras P, Cook JL, Kent PM. Anthropometric risk factors<br />
for patellar tendon injury among volleyball players. Br J<br />
Sports Med 2007;41:259�63.<br />
14. Skrzynski S. DSC study of collagen in disc disease. J Biophys<br />
2009;2009:819635, dx.doi.org/10.1155/2009/819635.<br />
15. Luevano-Contreras C, Chapman-Novakofski K. Dietary<br />
advanced glycation end products and aging. Nutrients<br />
2010;2(12):1247�65 [2009;2009:819635].<br />
16. Abate M, Schiavone C, Pelotti P, Salini V. Limited joint<br />
mobility (LJM) in elderly subjects with type II diabetes<br />
mellitus. Arch Gerontol Geriatrics 2011;53:135�40.<br />
17. Sakellaridis N. The influence of diabetes mellitus on lumbar<br />
intervertebral disk herniation. Surg Neurol 2006;66:152�4.<br />
18. Shepherd PR, Kahn BB. Glucose transporters and insulin<br />
action: implications for insulin resistance and diabetes<br />
mellitus. New Eng J Med 1999;341(4):248�57.<br />
19. Abdul-Ghani MA, DeFronzo RA. Pathogenesis of insulin<br />
resistance in skeletal muscle. J Biomed Biotechnol 2010:19,<br />
dx.doi.org/10.1155/2010/476279 [Article ID 476279].<br />
20. [Internet]American Heart Association. About metabolic<br />
syndrome. Dallas: The Association; 2014. [Available from<br />www.heart.org/HEARTORG/Conditions/More/<br />MetabolicSyndrome/About-Metabolic-Syndrome_UCM_<br />301920_Article.jsp].<br />
21. Hotamisligil GS. Inflammation and metabolic disorders.<br />
Nature 2006;444:860�7.<br />
22. Glass CK, Olefsky JM. Inflammation and lipid signaling in the<br />
etiology of insulin resistance. Cell Metab 2012;15(5):635�45.<br />
23. Reaven GM. All obese individuals are not created equal:<br />
insulin resistance is the major determinant of cardiovascular<br />
disease in overweight/obese individuals. Diabetes Vasc Dis<br />
Res 2005;2:105�12.<br />
24. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster<br />
BH, Kelley DE. Deficiency of subsarcolemmal mitochondria<br />
in obesity and type 2 diabetes. Diabetes 2005;54:8�14.<br />
25. Corcoran MP, Lamon-Fava S, Fielding RA. Trans fats and<br />
insulin resistance: skeletal muscle lipid deposition and insulin<br />
resistance: effect of dietary fatty acids and exercise. Am J Clin<br />
Nutr 2007;85:662�77.<br />
26. Schipper HS, Prakken B, Kalkhoven E, Boes M. Adipose<br />
tissue-resident immune cells: key players in immunometabolism.<br />
Trends Endocrinol Metab 2012;23:407�15.<br />
27. Antuna-Puente B, Feve B, Fellahi S, Bastard JP. Adipokines:<br />
the missing link between insulin resistance and obesity.<br />
Diabetes Metab 2008;34:2�11.<br />
28. Grimble RF. Inflammatory status and insulin resistance. Curr<br />
Opin Clin Nutr Metab Care 2003;5:551�9.<br />
29. Tilg H, Moschen AR. Inflammatory mechanisms in<br />
the regulation of insulin resistance. Mol Med 2008;3�4:222�31.<br />
30. Johnson DR, O’Conner JC, Satpathy A, Freund GG.<br />
Cytokines in type 2 diabetes. Vitam Horm 2006;74:405�41.<br />
31. Ridker PM, Wilson PW, Grundy SM. Should C-reactive<br />
protein be added to the metabolic syndrome and to<br />
assessment of global cardiovascular risk? Circulation 2004;<br />
109:2818�25.<br />
32. Gelaye B, Revilla L, Lopez T, et al. Association between<br />
insulin resistance and c-reactive protein among Peruvian<br />
adults. Diabetol Metab Syn 2010;2:30.<br />
33. Singh VP, Bali A, Singh N, et al. Advanced glycation end<br />
products and diabetic complications. Korean J Physiol<br />
Pharmacol 2014;18(1):1�14.<br />
34. Baker RG, Hayden MS. NF-kB, inflammation and metabolic<br />
disease. Cell Metab 2011;13(1):11�22.<br />
35. Purkayastha S, Cair D. Neuroinflammatory basis of metabolic<br />
syndrome. Mol Metab Nov 2013;2(4):356�63.<br />
36. Ehse JA, Boni-Schnetzler M, Faulenbach M, Donath MY.<br />
Macrophages, cytokines and beta-cell death in type 2 diabetes.<br />
Biochem Soc Trans 2008;36(3):340�2.<br />
37. Boni-Schnetzler M, Ehses JA, Faulenbach M, Donath MY.<br />
Insulitis in type 2 diabetes. Diabetes Obes Metab 2008;10<br />
(Suppl 4):201�4.<br />
38. Donath MY, Schumann DM, Faulenbach M, Ellingsgaard H,<br />
Perren A, Ehses JA. Islet inflammation in type 2<br />
diabetes: from metabolic stress to therapy. Diabetes Care<br />
2008;31(Suppl 2):S161�4.<br />
39. Donath MY, Boni-Schnetzler M, Ellingsgaard H, Ehses JA.<br />
Islet inflammation impairs the pancreatic beta-cell in type 2<br />
diabetes. Physiology 2009;24:325�31.<br />
40. Harford KA, Reynolds CM, McGillicuddy FC, Roche HM.<br />
Fats, inflammation and insulin resistance: insights to the role<br />
of macrophage and T-cell accumulation in adipose tissue.<br />
Proc Nutr Soc 2011;70:408�17.<br />
41. Munoz A, Costa M. Nutritionally mediated oxidative stress and<br />
inflammation. Oxid Med Cell Longev 2013;2013:610950, <br />
dx.doi.org/10.1155/2013/610950.<br />
42. Wisse BE, Schwartz MW. Does hypothalamic inflammation<br />
cause obesity? Cell Metab 2009;10(4):241�2.<br />
43. Purkayastha S, Cair D. Neuroinflammatory basis of metabolic<br />
syndrome. Mol Metab Nov 2013;2(4):356�63.<br />
44. Calegari VC, Torsoni AS, Vanzela EC, Ara�jo EP, Morari<br />
J, Zoppi CC, et al. Inflammation of the hypothalamus leads<br />
to defective pancreatic islet function. J Biol Chem 2011;<br />
286(15):12870�80.<br />
45. Cordain L, Eaton SB, Sebastian A, et al. Origins and evolution<br />
of the Western diet: health implications for the 21st century.<br />
Am J Clin Nutr 2005;81:341�54.<br />
46. Barclay AW, Petocz P, McMillan-Price J, et al. Glycemic<br />
index, glycemic load, and chronic disease risk�a metaanalysis<br />
of observational studies. Am J Clin Nutr<br />
2008;87:627�37.<br />
47. Facchini FS, Hua N, Abbasi F, Reaven GM. Insulin resistance<br />
as a predictor of age-related disease. J Clin Endocrinol Metab<br />
2001;86:3574�8.<br />
48. Lin H, Lee B, Ho Y, et al. Postprandial glucose improves the<br />
risk prediction of cardiovascular death beyond the metabolic<br />
syndrome in the nondiabetic population. Diabetes Care Sep<br />
2009;32(9):1721�6.<br />
49. O’Keefe JH, Bell DS. Postprandial hyperglycemia/<br />
hyperlipidemia (postprandial dysmetabolism) is a cardiovascular<br />
risk factor. Am J Cardiol 2007;100:899�904.<br />
50. Cao H. Adipocytokines in obesity and metabolic disease.<br />
J Endocrinol 2014;220(2):T47�59.<br />
51. Nah SS, Choi IY, Lee CK, et al. Effects of advanced glycation<br />
end products on the expression of COX2, PGE2 and NO in human osteoarthritic chondrocytes. Rheumatology (Oxford)<br />
2008;47(4):425�31.<br />
52. Abate M, Schiavone C, Pelotti P, Salini V. Limited joint<br />
mobility (LJM) in elderly subjects with type II diabetes<br />
mellitus. Arch Gerontol Geriatr 2011;53:135�40.<br />
53. Robinson D, Mirovsky Y, Halperin N, Evron Z, Nevo Z.<br />
Changes in proteoglycans of intervertebral disc in diabetic<br />
patients: a possible cause of increased back pain. Spine<br />
1998;23:849�56.<br />
54. Sakellaridis N, Androulis A. Influence of diabetes mellitus on<br />
cervical intervertebral disc herniation. Clin Neurol Neurosurg<br />
2008;110:810�2.<br />
55. Jhawar BS, Fuchs CS, Colditz GA, Stampfer MJ. Cardiovascular<br />
risk factors for physician-diagnosed lumbar disc<br />
herniation. Spine J 2006;6:684�91.<br />
56. Lotan R, Oron A, Anekstein Y, Shalmon E, Mirovsky Y.<br />
Lumbar stenosis and systemic diseases: is there any relevance.<br />
J Spinal Disord Tech 2008;21:247�51.<br />
57. Anekstein Y, Smorgick Y, Lotan R, et al. Diabetes mellitus as<br />
a risk factor for the development of lumbar spinal stenosis. Isr<br />
Med Assoc J 2010;12:16�20.<br />
58. Choi KM. Sarcopenia and sarcopenic obesity. Endocrinol<br />
Metab (Seoul) 2013;28(2):86�9.<br />
59. D’hooge R, Cagnie B, Crombez G, et al. Increased<br />
intramuscular fatty infiltration without differences in lumbar<br />
muscle cross-sectional area during remission of unilateral<br />
recurrent low back pain. Man Ther 2012 Dec;17(6):5584�8.<br />
60. Chen YY, Pao JL, Liaw CK, et al. Image changes of paraspinal<br />
muscles and clinical correlations in patients with unilateral<br />
lumbar spinal stenosis. Eur Spine J 2014;23(5):999�1006.<br />
61. Kim Y, Park H. Does regular exercise without weight loss<br />
reduce insulin resistance in children and adolescents? In J<br />
Endocrinol 2013:402592, dx.doi.org/10.1155/2013/<br />
402592 [Epub 2013 Dec 12].<br />
62. Strasser B, Siebert U, Schobersberger W. Resistance training<br />
in the treatment of the metabolic syndrome: a systematic<br />
review and meta-analysis of the effect of resistance training on<br />
metabolic clustering in patients with abnormal glucose<br />
metabolism. Sports Med 2010;40:397�415.<br />
63. Sharman MJ, Volek JS. Weight loss leads to reductions in<br />
inflammatory biomarkers after a very-low-carbohydrate diet<br />
and a low-fat diet in overweight men. Clin Sci (Lond)<br />
2004;13:365�9.<br />
64. Teng KT, Chang CY, Chang LF, et al. Modulation of obesityinduced<br />
inflammation by dietary fats: mechanisms and<br />
clinical evidence. Nutr J 2014;13:12, dx.doi.org/<br />
10.1186/1475-2891-13-12.<br />
65. Tzotzas T, Evangelou P, Kiortsis DN. Obesity, weight loss<br />
and conditional cardiovascular risk factors. Obes Rev 2011;12<br />
(5):e282�9.<br />
66. Stone N, Robinson J, Lichtenstein AH, et al. 2013 ACC/AHA<br />
Guideline on the Treatment of Blood Cholesterol to Reduce<br />
Atherosclerotic Cardiovascular Risk in Adults: A report of<br />
the American College of Cardiology/American Heart<br />
Association Task Force on practice guidelines. Circulation<br />
2014;129(25 Suppl 2):S1�S45.<br />
67. P�rez-Guisado J, Mu�oz-Serrano A. A pilot study of the<br />
Spanish ketogenic Mediterranean diet: an effective therapy for<br />
the metabolic syndrome. J Med Food 2011;14(7�8):681�7.<br />
68. P�rez-Guisado J, Mu�oz-Serrano A, Alonso-Moraga A.<br />
Spanish ketogenic Mediterranean diet: a healthy cardiovascular<br />
diet for weight loss. Nutr J 2008;7:30, dx.doi.org/<br />
10.1186/1475-2891-7-30.<br />
69. Jonsson T, Granfeldt Y, Lindeberg S, et al. Subjective satiety<br />
and other experiences of a Paleolithic diet compared to a<br />
diabetes diet in patients with T2DM. Nutr J 2013;12:105,<br />
dx.doi.org/10.1186/1475-2891-12-105.<br />
70. Jonsson T, Granfeldt Y, Ahren B, et al. Beneficial effects of a<br />
Paleolithic diet on cardiovascular risk factors in T2DM: a<br />
randomized cross-over pilot study. Cardiovasc Diabetol<br />
2009;8:35, dx.doi.org/10.1186/1475-2840-8-35.<br />
71. Nicklas BJ, You T, Pahor M. Behavioural treatments<br />
for chronic system inflammation: effects of dietary<br />
weight loss and exercise training. Can Med Assoc J<br />
2005;172(9):1199�209.<br />
72. O’Keefe JH, Gheewala NM, O’Keefe JO. Dietary<br />
strategies for improving post-prandial glucose, lipids, inflammation,<br />
and cardiovascular health. J Am Coll Cardiol<br />
2008;51:249�55.<br />
73. O’Keefe Jr JH, Cordain L. Cardiovascular disease resulting<br />
from a diet and lifestyle at odds with our Paleolithic genome:<br />
how to become a 21st-century hunter�gatherer. Mayo Clin<br />
Proc 2004;79(1):101�8.<br />
74. Ames BN. Low micronutrient intake may accelerate the<br />
degenerative diseases of aging through allocation of scarce<br />
micronutrients by triage. Proc Natl Acad Sci U S A 2006;103<br />
(47):17589�94.<br />
75. Holick MF, Chen TC. Vitamin D deficiency: a worldwide<br />
problem with health consequences. Am J Clin Nutr<br />
2008;87:1080S�6S [Suppl.].<br />
76. Toubi E, Shoenfeld Y. The role of vitamin D in regulating<br />
immune responses. Isr Med Assoc J 2010;12(3):174�5.<br />
77. King DE, Mainous AG, Geesey ME, Egan BM, Rehman S.<br />
Magnesium supplement intake and C-reactive protein levels<br />
in adults. Nutr Res 2006;26:193�6.<br />
78. Rosanoff A, Weaver CM, Rude RK. Suboptimal magnesium<br />
status in the United States: are the health consequences<br />
underestimated? Nutr Rev 2012;70(3):153�64.<br />
79. Simopoulos AP. Omega-3 fatty acids in inflammation and<br />
autoimmune diseases. J Am Coll Nutr 2002;21(6):495�505.<br />
80. Simopoulos AP. The importance of the omega-6/omega-3<br />
fatty acid ratio in cardiovascular disease and other chronic<br />
diseases. Exp Biol Med 2008;233:674�88.<br />
81. Fung GJ, Steffen LM, Zhou X, et al. Vitamin D intake is<br />
inversely related to risk of developing metabolic syndrome<br />
in African American and white men and women over 20 y:<br />
the Coronary Artery Risk Development in Young Adults<br />
study. Am J Clin Nutr 2012;96(1):24�9 [Published online<br />2012 May 30].<br />
82. Palomer X, Gonzalez-Clemente JM, Blanco-Vaca F, Mauricio<br />
D. Role of vitamin D in the pathogenesis of type 2 diabetes<br />
mellitus. Diabetes Obes Metab 2008;10:185�97.<br />
83. Guadarrama-Lopez AL, Valdes-Ramos R, Martinex-Carrillo<br />
BE. T2DM, PUFAs, and vitamin D: their relation to<br />
inflammation. J Immunol Res 2014;2014:860703, dx.<br />
doi.org/10.1155/2014/860703.<br />
84. Cannell JJ, Hollis BW. Use of vitamin D in clinical practice.<br />
Altern Med Rev 2008;13(1):6�20.<br />
85. Davidson MB, Duran P, Lee ML, Friedman TC. High-dose<br />
vitamin D supplementation in people with prediabetes and<br />
hypovitaminosis D. Diabetes Care 2013;36(2):260�6, <br />
dx.doi.org/10.2337/dc12-1204.<br />
86. Schwalfenberg G. Vitamin D, and diabetes: improvement of<br />
glycemic control with vitamin D3 repletion. Can Fam<br />
Physician 2008;54:864�6.<br />
87. Kim DJ, Xun P, Liu K, et al. Magnesium intake in relation to<br />
systemic inflammation, insulin resistance, and the incidence<br />
of diabetes. Diabetes Care 2010;33(12):2604�10, dx.<br />
doi.org/10.2337/dc10-0994.<br />
88. Guerrero-Romero F, Tamez-Perez HE, Gonz�lez-Gonz�lez G,<br />
et al. Oral magnesium supplementation improves insulin<br />
sensitivity in non-diabetic subjects with insulin resistance. A<br />
double-blind placebo-controlled randomized trial. Diabetes<br />
Metab 2004;30(3):253�8.<br />
89. Rodr�guez-Mor�n M, Guerrero-Romero F. Oral magnesium<br />
supplementation improves insulin sensitivity and metabolic<br />
control in type 2 diabetic subjects: a randomized double-blind<br />
controlled trial. Diabetes Care 2003;26(4):1147�52.<br />
90. Song Y, He K, Levitan EB, Manson JE, Liu S. Effects of oral<br />
magnesium supplementation on glycaemic control in type 2<br />
diabetes: a meta-analysis of randomized double-blind controlled<br />
trials. Diabet Med 2006;23(10):1050�6.<br />
91. Mooren FC, Kr�ger K, V�lker K, Golf SW,Wadepuhl M, Kraus<br />
A. Oral magnesium supplementation reduces insulin resistance<br />
in non-diabetic subjects�a double-blind, placebo-controlled,<br />
randomized trial. Diabetes Obes Metab 2011;13(3):281�4.<br />
92. Aggarwal BB. Targeting inflammation induced obesity and<br />
metabolic diseases by curcumin and other nutraceuticals.<br />
Annu Rev Nutr 2010;30:173�9.<br />
93. Alappat L, Awad AB. Curcumin and obesity: evidence and<br />
mechanisms. Nutr Rev 2010;68(12):729�38.<br />
94. Gonzales AM, Orlando RA. Curcumin and resveratrol inhibit<br />
nuclear factor-kappaB-mediated cytokine expression in adipocytes.<br />
Nutr Metab 2008;5:17, dx.doi.org/10.1186/<br />
1743-7075-5-17.<br />
95. Sahebkar A. Why it is necessary to translate curcumin into<br />
clinical practice for the prevention and treatment of metabolic<br />
syndrome? Biofactors 2012, dx.doi.org/10.1002/<br />
biof.1062 [Epub ahead of print].<br />
96. Hsu CH, Cheng AL. Clinical studies with curcumin. Adv Exp<br />
Med Biol 2007;595:471�80.<br />
97. Chuengsamarn S, Rattanamongkolgul S, Luechapudiporn R,<br />
Phisalaphong C, Jirawatnotai S. Curcumin extract for prevention<br />
of type 2 diabetes. Diabetes Care 2012;35(11):2121�7.<br />
98. Jurenka JS. Anti-inflammatory properties of curcumin, a<br />
major constituent of curcuma longa: a review of preclinical<br />
and clinical research. Altern Med Rev 2009;14(2):141�53.<br />
99. Leach M. Gymnema sylvestre for diabetes mellitus: a systematic<br />
review. J Altern Complement Med 2007;13(9):977�83.<br />
100. Chattopadhyay R. A comparative evaluation of some blood<br />
sugar lowering agents of plant origin. J Ethnopharmacol<br />
1999;67:367�72.<br />
101. Nahas R, Moher M. Complementary and alternative medicine<br />
for the treatment of type 2 diabetes. Can Fam Physician<br />
2009;55:591�6.<br />
102. Vanadium/Vanadyl sulfate: monograph. Altern Med Rev<br />
2009;14:17�80.<br />
103. Boden G, Chen X, Ruiz J, et al. Effects of vanadyl sulfate<br />
on carbohydrate and lipid metabolism in patients with noninsulin-dependent<br />
diabetes mellitus. Metabolism 1996;45:<br />
1130�5.<br />
104. Jacques-Camarena O, Gonz�lez-Ortiz M, Mart�nez-Abundis E,<br />
et al. Effect of vanadium on insulin sensitivity in patients with<br />
impaired glucose tolerance. Ann Nutr Metab 2008;53:195�8.<br />
105. Vincent JB. The biochemisty of chromium. J Nutr<br />
2000;130:715�8.<br />
106. Anderson RA. Chromium and insulin resistance. Nutr Res<br />
Rev 2003;16:267�75.<br />
107. Vincent JB. Chromium: celebrating 50 years as an essential<br />
element? Dalton Trans 2010;39:3787�94.<br />
108. Office of Dietary Supplements. [Internet]. Dietary supplement<br />
fact sheet: Chromium. Washington, DC: United States<br />
Department of Health and Human Services. ods.od.nih.<br />
gov/factsheets/chromium/. Reviewed November 4, 2013.<br />
109. Anderson RA. Chromium, glucose intolerance and diabetes.<br />
J Am Coll Nutr 1998;17(6):548�55.<br />
110. Cefalu WT, Rood J, Patricia Pinsonat P, et al. Characterization<br />
of the metabolic and physiologic response to chromium<br />
supplementation in subjects with type 2 diabetes mellitus.<br />
Metab Clin Exp 2010;59:755�62.<br />
111. Heimbach JT, Anderson RA. Chromium: recent studies regarding<br />
nutritional roles and safety. Nutr Today 2005;40(4):180�95.<br />
112. Shay KP, Moreau RF, Smith EJ, Smith AR, Hagen TM.<br />
Alpha-lipoic acid as a dietary supplement: molecular<br />
mechanisms and therapeutic potential. Biochim Biophys<br />
Acta 2009;1790:1149�60.<br />
113. Morikawa T, Yasuno R, Wada H. Do mammalian cells<br />
synthesize lipoic acid? Identification of a mouse cDNA<br />
encoding a lipoic acid synthase located in mitochondria.<br />
FEBS Lett 2001;498:16�21.<br />
114. Singh U, Jialal I. Alpha-lipoic acid supplementation and<br />
diabetes. Nutr Rev 2008;66(11):646�57.<br />
115. Padmalayam I, Hasham S, Saxena U, Pillarisetti S. Lipoic acid<br />
synthase (LASY): a novel role in inflammation, mitochondrial<br />
function, and insulin resistance. Diabetes 2009;58:600�8.<br />
116. Capasso I, Esposito E, Maurea N, et al. Combination of<br />
inositol and alpha lipoic acid in metabolic syndrome-affected<br />
women: a randomized placebo-controlled trial. Trial<br />
2013;14:273, dx.doi.org/10.1186/1745-6215-14-273.<br />
117. Udupa A, Nahar P, Shah S, et al. A comparative study of<br />
effects of omega-3 fatty acids, alpha lipoic acid and vitamin E<br />
in T2DM. Ann Med Health Sci Res 2013;3(3):442�6.

Close Accordion
The Role Of Epigenetics In Obesity And Metabolic Disease

The Role Of Epigenetics In Obesity And Metabolic Disease

Epigenetic Abstract:

The increased prevalence of obesity and related comorbidities is a major public health problem. While genetic factors undoubtedly play a role in determining individual susceptibility to weight gain and obesity, the identified genetic variants only explain part of the variation. This has led to growing interest in understanding the potential role of epigenetics as a mediator of gene-environment interactions underlying the development of obesity and its associated comorbidities. Initial evidence in support of a role of epigenetics in obesity and type 2 diabetes mellitus (T2DM) was mainly provided by animal studies, which reported epigenetic changes in key metabolically important tissues following high-fat feeding and epigenetic differences between lean and obese animals and by human studies which showed epigenetic changes in obesity and T2DM candidate genes in obese/diabetic individuals. More recently, advances in epigenetic methodologies and the reduced cost of epigenome-wide association studies (EWAS) have led to a rapid expansion of studies in human populations. These studies have also reported epigenetic differences between obese/T2DM adults and healthy controls and epigenetic changes in association with nutritional, weight loss, and exercise interventions. There is also increasing evidence from both human and animal studies that the relationship between perinatal nutritional exposures and later risk of obesity and T2DM may be mediated by epigenetic changes in the offspring. The aim of this review is to summarize the most recent developments in this rapidly moving field, with a particular focus on human EWAS and studies investigating the impact of nutritional and lifestyle factors (both pre- and postnatal) on the epigenome and their relationship to metabolic health outcomes. The difficulties in distinguishing consequence from causality in these studies and the critical role of animal models for testing causal relationships and providing insight into underlying mechanisms are also addressed. In summary, the area of epigenetics and metabolic health has seen rapid developments in a short space of time. While the outcomes to date are promising, studies are ongoing, and the next decade promises to be a time of productive research into the complex interactions between the genome, epigenome, and environment as they relate to metabolic disease.

Keywords: Epigenetics, DNA methylation, Obesity, Type 2 diabetes, Developmental programming

Introduction

Epigenetic mechanismsObesity is a complex, multifactorial disease, and better understanding of the mechanisms underlying the interactions between lifestyle, environment, and genetics is critical for developing effective strategies for prevention and treatment [1].

In a society where energy-dense food is plentiful and the need for physical activity is low, there is a wide variation in individuals� susceptibility to develop�obesity and metabolic health problems. Estimates of the role of heredity in this variation are in the range of 40�70 %, and while large genome-wide association studies (GWAS) have identified a number of genetic loci associated with obesity risk, the ~100 most common genetic variants only account for a few percent of variance in obesity [2, 3]. Genome-wide estimates are higher, accounting for ~20 % of the variation [3]; however, a large portion of the heritability remains unexplained.

Recently, attention has turned to investigating the role of epigenetic changes in the etiology of obesity. It has been argued that the epigenome may represent the mechanistic link between genetic variants and environmental�factors in determining obesity risk and could help explain the �missing heritability.� The first human epigenetic studies were small and only investigated a limited number of loci. While this generally resulted in poor reproducibility, some of these early findings, for instance the relationship between PGC1A methylation and type 2 diabetes mellitus (T2DM) [4] and others as discussed in van Dijk et al. [5], have been replicated in later studies. Recent advances and increased affordability of high- throughput technologies now allow for large-scale epigenome wide association studies (EWAS) and integration of different layers of genomic information to explore the complex interactions between the genotype, epigenome, transcriptome, and the environment [6�9]. These studies are still in their infancy, but the results thus far have shown promise in helping to explain the variation in obesity susceptibility.

There is increasing evidence that obesity has develop mental origins, as exposure to a suboptimal nutrient supply before birth or in early infancy is associated with an increased risk of obesity and metabolic disease in later life [10�13]. Initially, animal studies demonstrated that a range of early life nutritional exposures, especially those experienced early in gestation, could induce epigenetic changes in key metabolic tissues of the offspring that persisted after birth and result in permanent alterations in gene function [13�17]. Evidence is emerging to support the existence of the same mechanism in humans. This has led to a search for epigenetic marks present early in life that predict later risk of metabolic disease, and studies to determine whether epigenetic programming of metabolic disease could be prevented or reversed in later life.

This review provides an update of our previous systematic review of studies on epigenetics and obesity in humans [5]. Our previous review showcased the promising outcomes of initial studies, including the first potential epigenetic marks for obesity that could be detected at birth (e.g., RXRA) [18]. However, it also highlighted the limited reproducibility of the findings and the lack of larger scale longitudinal investigations. The current review focuses on recent developments in this rapidly moving field and, in particular, on human EWAS and studies investigating the impact of (pre- and postnatal) nutritional and lifestyle factors on the epigenome and the emerging role of epigenetics in the pathology of obesity. We also address the difficulties in identifying causality in these studies and the importance of animal models in providing insight into mechanisms.

Review

Epigenetic Changes In Animal Models Of Obesity

rabbit eatingAnimal models provide unique opportunities for highly controlled studies that provide mechanistic insight into�the role of specific epigenetic marks, both as indicators of current metabolic status and as predictors of the future risk of obesity and metabolic disease. A particularly important aspect of animal studies is that they allow for the assessment of epigenetic changes within target tissues, including the liver and hypothalamus, which is much more difficult in humans. Moreover, the ability to harvest large quantities of fresh tissue makes it possible to assess multiple chromatin marks as well as DNA methylation. Some of these epigenetic modifications either alone or in combination may be responsive to environmental programming. In animal models, it is also possible to study multiple generations of offspring and thus enable differentiation between trans-generational and intergenerational transmission of obesity risk mediated by epigenetic memory of parental nutritional status, which cannot be easily distinguished in human studies. We use the former term for meiotic transmission of risk in the absence of continued exposure while the latter primarily entails direct transmission of risk through metabolic reprogramming of the fetus or gametes.

Animal studies have played a critical role in our current understanding of the role of epigenetics in the developmental origins of obesity and T2DM. Both increased and decreased maternal nutrition during pregnancy have been associated with increased fat deposition in offspring of most mammalian species studied to date (reviewed in [11, 13�15, 19]). Maternal nutrition during pregnancy not only has potential for direct effects on the fetus, it also may directly impact the developing oocytes of female fetuses and primordial germ cells of male fetuses and therefore could impact both the off- spring and grand-offspring. Hence, multigenerational data are usually required to differentiate between maternal intergenerational and trans-generational transmission mechanisms.

Table 1 summarizes a variety of animal models that have been used to provide evidence of metabolic and epigenetic changes in offspring associated with the parental plane of nutrition. It also contains information pertaining to studies identifying altered epigenetic marks in adult individuals who undergo direct nutritional challenges. The table is structured by suggested risk transmission type.

table 1(i) Epigenetic Changes In Offspring Associated With Maternal Nutrition During Gestation

Maternal nutritional supplementation, undernutrition, and over nutrition during pregnancy can alter fat deposition and energy homeostasis in offspring [11, 13�15, 19]. Associated with these effects in the offspring are changes in DNA methylation, histone post-translational modifications, and gene expression for several target genes,�especially genes regulating fatty acid metabolism and insulin signaling [16, 17, 20�30]. The diversity of animal models used in these studies and the common metabolic pathways impacted suggest an evolutionarily conserved adaptive response mediated by epigenetic modification. However, few of the specific identified genes and epigenetic changes have been cross-validated in related studies, and large-scale genome-wide investigations have typically not been applied. A major hindrance to comparison of these studies is the different develop mental windows subjected to nutritional challenge, which may cause considerably different outcomes. Proof that the epigenetic changes are causal rather than being associated with offspring phenotypic changes is also required. This will necessitate the identification of a parental nutritionally induced epigenetic �memory� response that precedes development of the altered phenotype in offspring.

(ii)Effects Of Paternal Nutrition On Offspring Epigenetic Marks

baby sleeping holding handsEmerging studies have demonstrated that paternal plane of nutrition can impact offspring fat deposition and epigenetic marks [31�34]. One recent investigation using mice has demonstrated that paternal pre-diabetes leads to increased susceptibility to diabetes in F1 offspring with associated changes in pancreatic gene expression and DNA methylation linked to insulin signaling [35]. Importantly, there was an overlap of these epigenetic changes in pancreatic islets and sperm suggesting germ line inheritance. However, most of these studies, although intriguing in their implications, are limited in the genomic scale of investigation and frequently show weak and somewhat transient epigenetic alterations associated with mild metabolic phenotypes in offspring.

(iii)Potential Trans-generational Epigenetic Changes Promoting Fat Deposition In Offspring

excess nutritionStable transmission of epigenetic information across multiple generations is well described in plant systems and C. elegans, but its significance in mammals is still much debated [36, 37]. An epigenetic basis for grand- parental transmission of phenotypes in response to dietary exposures has been well established, including in livestock species [31]. The most influential studies demonstrating effects of epigenetic transmission impacting offspring phenotype have used the example of the viable yellow agouti (Avy) mouse [38]. In this mouse, an insertion of a retrotransposon upstream of the agouti gene causes its constitutive expression and consequent yellow coat color and adult onset obesity. Maternal transmission through the germ line results in DNA methylation�mediated silencing of agouti expression resulting in wild-type coat color and lean phenotype of the offspring [39, 40]. Importantly, subsequent studies in these mice demonstrated that maternal exposure to methyl donors causes a shift in coat color [41]. One study has reported transmission of a phenotype to the F3 generation and alterations in expression of large number of genes in response to protein restriction in F0 [42]; however, alterations in expression were highly variable and a direct link to epigenetic changes was not identified in this system.

(iv) Direct Exposure Of Individuals To Excess Nutrition In Postnatal Life

modern western lifestyleWhile many studies have identified diet-associated epigenetic changes in animal models using candidate site-specific regions, there have been few genome-wide analyses undertaken. A recent study focussed on determining the direct epigenetic impact of high-fat diets/ diet-induced obesity in adult mice using genome-wide gene expression and DNA methylation analyses [43]. This study identified 232 differentially methylated regions (DMRs) in adipocytes from control and high-fat fed mice. Importantly, the corresponding human regions for the murine DMRs were also differentially methylated in adipose tissue from a population of obese and lean humans, thereby highlighting the remarkable evolutionary conservation of these regions. This result emphasizes the likely importance of the identified DMRs in regulating energy homeostasis in mammals.

Human Studies

anatomy 3D model

Drawing on the evidence from animal studies and with the increasing availability of affordable tools for genome- wide analysis, there has been a rapid expansion of epigenome studies in humans. These studies have mostly focused on the identification of site-specific differences in DNA methylation that are associated with metabolic phenotypes.

A key question is the extent to which epigenetic modifications contribute to the development of the metabolic phenotype, rather than simply being a con- sequence of it (Fig. 1). Epigenetic programming could contribute to obesity development, as well as playing a role in consequent risk of cardiovascular and metabolic problems. In human studies, it is difficult to prove causality [44], but inferences can be made from a number of lines of evidence:

fig 1(i) Genetic association studies. Genetic polymorphisms that are associated with an increased risk of developing particular conditions are a priori linked to the causative genes. The presence of differential�methylation in such regions infers functional relevance of these epigenetic changes in controlling expression of the proximal gene(s). There are strong cis-acting genetic effects underpinning much epigenetic variation [7, 45], and in population-based studies, methods that use genetic surrogates to infer a causal or mediating role of epigenome differences have been applied [7, 46�48]. The use of familial genetic information can also lead to the identification of potentially causative candidate regions showing phenotype-related differential methylation [49].

(ii)Timing of epigenetic changes. The presence of an epigenetic mark prior to development of a phenotype is an essential feature associated with causality. Conversely, the presence of a mark in association with obesity, but not before its development, can be used to exclude causality but would not exclude a possible role in subsequent obesity-related pathology.

(iii)Plausible inference of mechanism. This refers to epigenetic changes that are associated with altered expression of genes with an established role in regulating the phenotype of interest. One such example is the association of methylation at two CpG sites at the CPT1A gene with circulating triglyceride levels [50]. CPT1A encodes carnitine palmitoyltransferase 1A, an enzyme with a central role in fatty acid metabolism, and this is strongly indicative that differential methylation of this gene may be causally related to the alterations in plasma triglyceride concentrations.

Epigenome-Wide Association Studies: Identifying Epigenetic Biomarkers Of Metabolic Health

A number of recent investigations have focused on exploring associations between obesity/metabolic diseases�and DNA methylation across the genome (Table 2). The largest published EWAS so far, including a total of 5465 individuals, identified 37 methylation sites in blood that were associated with body mass index (BMI), including sites in CPT1A, ABCG1, and SREBF1 [51]. Another large-scale study showed consistent associations between BMI and methylation in HIF3A in whole blood and adipose tissue [52], a finding which was also partially replicated in other studies [9, 51]. Other recently reported associations between obesity-related measures and DNA methylation include (i) DNA methylation differences between lean and obese individuals in LY86 in blood leukocytes [53]; (ii) associations between PGC1A promoter methylation in whole blood of children and adiposity 5 years later [54]; (iii) associations between waist-hip ratio and ADRB3 methylation in blood [55]; and (iv) associations between BMI, body fat distribution measures, and multiple DNA methylation sites in adipose tissue [9, 56]. EWAS have also shown associations between DNA methylation sites and blood lipids [55, 57�59], serum metabolites [60], insulin resistance [9, 61], and T2DM [48, 62, 63] (Table 2).

table 2 contdFrom these studies, altered methylation of PGC1A, HIF3A, ABCG1, and CPT1A and the previously described RXRA [18] have emerged as biomarkers associated with, or perhaps predictive of, metabolic health that are also plausible candidates for a role in development of metabolic disease.

Interaction Between Genotype And The Epigenome

Genotype EpigenomeEpigenetic variation is highly influenced by the underlying genetic variation, with genotype estimated to explain ~20�40 % of the variation [6, 8]. Recently, a number of studies have begun to integrate methylome and genotype data to identify methylation quantitative trait loci (meQTL) associated with disease phenotypes. For instance, in adipose tissue, an meQTL overlapping�with a BMI genetic risk locus has been identified in an enhancer element upstream of ADCY3 [8]. Other studies have also identified overlaps between known obesity and T2DM risk loci and DMRs associated with obesity and T2DM [43, 48, 62]. Methylation of a number of such DMRs was also modulated by high-fat feeding in mice [43] and weight loss in humans [64]. These results identify an intriguing link between genetic variations linked with disease susceptibility and their association with regions of the genome that undergo epigenetic modifications in response to nutritional challenges, implying a causal relationship. The close connection between genetic and epigenetic variation may signify their essential roles in generating individual variation [65, 66]. However, while these findings suggest that DNA methylation may be a mediator of genetic effects, it is also important to consider that both genetic and epigenetic processes could act independently on the same genes. Twin studies [8, 63, 67] can provide important insights and indicate that inter-individual differences in levels of DNA methylation arise predominantly from non-shared environment and stochastic influences, minimally from shared environmental effects, but also with a significant impact of genetic variation.

The Impact Of The Prenatal And Postnatal Environment On The Epigenome

fetus modelPrenatal environment: Two recently published studies made use of human populations that experienced �natural� variations in nutrient supply to study the impact of maternal nutrition before or during pregnancy on DNA methylation in the offspring [68, 69]. The first study used a Gambian mother-child cohort to show that both seasonal variations in maternal methyl donor intake during pregnancy and maternal pre-pregnancy BMI were associated with altered methylation in the infants [69]. The second study utilized adult offspring from the Dutch Hunger Winter cohort to investigate the effect of prenatal exposure to an acute period of severe maternal undernutrition on DNA methylation of genes involved in growth and metabolism in adulthood [68]. The results highlighted the importance of the timing of the exposure in its impact on the epigenome, since significant epigenetic effects were only identified in individuals exposed to famine during early gestation. Importantly, the epigenetic changes occurred in conjunction with increased BMI; however, it was not possible to establish in this study whether these changes were present earlier in life or a consequence of the higher BMI.

Other recent studies have provided evidence that prenatal over-nutrition and an obese or diabetic maternal environment are also associated with DNA methylation changes in genes related to embryonic development, growth, and metabolic disease in the offspring [70�73].

While human data are scarce, there are indications that paternal obesity can lead to altered methylation of imprinted genes in the newborn [74], an effect thought to be mediated via epigenetic changes acquired during spermatogenesis.

baby walking in the grass and mudPostnatal environment: The epigenome is established de novo during embryonic development, and therefore, the prenatal environment most likely has the most significant impact on the epigenome. However, it is now clear that changes do occur in the �mature� epigenome under the influence of a range of conditions, including aging, exposure to toxins, and dietary alterations. For example, changes in DNA methylation in numerous genes in skeletal muscle and PGC1A in adipose tissue have been demonstrated in response to a high-fat diet [75, 76]. Interventions to lose body fat mass have also been associated with changes in DNA methylation. Studies have reported that the DNA methylation profiles of adipose tissue [43, 64], peripheral blood mononuclear cells [77], and muscle tissue [78] in formerly obese patients become more similar to the profiles of lean subjects following weight loss. Weight loss surgery also partially reversed non-alcoholic fatty liver disease-associated methylation changes in liver [79] and in another study led to hypomethylation of multiple obesity candidate genes, with more pronounced effects in subcutaneous compared to omental (visceral) fat [64]. Accumulating evidence suggests that exercise interventions can also influence DNA methylation. Most of these studies have been conducted in lean individuals [80�82], but one exercise study in obese T2DM subjects also demonstrated changes in DNA methylation, including in genes involved in fatty acid and glucose transport [83]. Epigenetic changes also occur with aging, and recent data suggest a role of obesity in augmenting them [9, 84, 85]. Obesity accelerated the epigenetic age of liver tissue, but in contrast to the findings described above, this effect was not reversible after weight loss [84].

Collectively, the evidence in support of the capacity to modulate the epigenome in adults suggests that there may be the potential to intervene in postnatal life to modulate or reverse adverse epigenetic programming.

Effect Sizes And Differences Between Tissue Types

connective tissuesDNA methylation changes associated with obesity or induced by diet or lifestyle interventions and weight loss are generally modest (<15 %), although this varies depending on the phenotype and tissue studied. For instance, changes greater than 20 % have been reported in adipose tissue after weight loss [64] and associations between HIF3A methylation and BMI in adipose tissue were more pronounced than in blood [52].

The biological relevance of relatively small methylation changes has been questioned. However, in tissues consisting of a mixture of cell types, a small change in DNA methylation may actually reflect a significant change in a specific cell fraction. Integration of epigenome data with transcriptome and other epigenetic data, such as histone modifications, is important, since small DNA methylation changes might reflect larger changes in chromatin structure and could be associated with broader changes in gene expression. The genomic context should also be considered; small changes within a regulatory element such as a promotor, enhancer, or insulator may have functional significance. In this regard, DMRs for obesity, as well as regions affected by prenatal famine exposure and meQTL for metabolic trait loci have been observed to overlap enhancer elements [8, 43, 68]. There is evidence that DNA methylation in famine-associated regions could indeed affect enhancer activity [68], supporting a role of nutrition-induced methylation changes in gene regulation.

A major limitation in many human studies is that epigenetic marks are often assessed in peripheral blood, rather than in metabolically relevant tissues (Fig. 2). The heterogeneity of blood is an issue, since different cell populations have distinct epigenetic signatures, but algorithms have been developed to estimate the cellular composition to overcome this problem [86]. Perhaps more importantly, epigenetic marks in blood cells may not necessarily report the status of the tissues of primary interest. Despite this, recent studies have provided clear evidence of a relationship between epigenetic marks in blood cells and BMI. In the case of HIF3A for which the level of methylation (beta-value) in the study population ranged from 0.14�0.52, a 10 % increase in methylation was associated with a BMI increase of 7.8 %�[52]. Likewise, a 10 % difference in PGC1A methylation may predict up to 12 % difference in fat mass [54].

fig 2Conclusions

The study of the role of epigenetics in obesity and metabolic disease has expanded rapidly in recent years, and evidence is accumulating of a link between epigenetic modifications and metabolic health outcomes in humans. Potential epigenetic biomarkers associated with obesity and metabolic health have also emerged from recent studies. The validation of epigenetic marks in multiple cohorts, the fact that several marks are found in genes with a plausible function in obesity and T2DM development, as well as the overlap of epigenetic marks with known obesity and T2DM genetic loci strengthens the evidence that these associations are real. Causality has so far been difficult to establish; however, regardless of whether the associations are causal, the identified epigenetic marks may still be relevant as biomarkers for obesity and metabolic disease risk.

Effect sizes in easily accessible tissues such as blood are small but do seem reproducible despite variation in ethnicity, tissue type, and analysis methods [51]. Also, even small DNA methylation changes may have biological significance. An integrative �omics� approach will be crucial in further unraveling the complex interactions between the epigenome, transcriptome, genome, and metabolic health. Longitudinal studies, ideally spanning multiple generations, are essential to establishing causal relationships. We can expect more such studies in the future, but this will take time.

While animal studies continue to demonstrate an effect of early life nutritional exposure on the epigenome and metabolic health of the offspring, human data are still limited. However, recent studies have provided clear�evidence that exposure to suboptimal nutrition during specific periods of prenatal development is associated with methylation changes in the offspring and therefore have the potential to influence adult phenotype. Animal studies will be important to verify human findings in a more controlled setting, help determine whether the identified methylation changes have any impact on metabolic health, and unravel the mechanisms underlying this intergenerational/transgenerational epigenetic regulation. The identification of causal mechanisms underlying metabolic memory responses, the mode of transmission of the phenotypic effects into successive generations, the degree of impact and stability of the transmitted trait, and the identification of an overarching and unifying evolutionary context also remain important questions to be addressed. The latter is often encapsulated by the predictive adaptive response hypothesis, i.e., a response to a future anticipated environment that increases fitness of the population. However, this hypothesis has increasingly been questioned as there is limited evidence for increased fitness later in life [87].

In summary, outcomes are promising, as the epigenetic changes are linked with adult metabolic health and they act as a mediator between altered prenatal nutrition and subsequent increased risk of poor metabolic health outcomes. New epigenetic marks have been identified that are associated with measures of metabolic health. Integration of different layers of genomic information has added further support to causal relationships, and there have been further studies showing effects of pre- and postnatal environment on the epigenome and health. While many important questions remain, recent methodological advances have enabled the types of large-scale population-based studies that will be required to address the knowledge gaps. The next decade promises to be a period of major activity in this important research area.

Susan J. van Dijk1, Ross L. Tellam2, Janna L. Morrison3, Beverly S. Muhlhausler4,5� and Peter L. Molloy1*�

Competing interests

The authors declare that they have no competing interests.

Authors� contributions
All authors contributed to the drafting and critical revision of the manuscript, and all authors read and approved the final manuscript.

Authors� information
Beverly S. Muhlhausler and Peter L. Molloy are joint last authors.

Acknowledgements

This work has been supported by a grant from the Science and Industry Endowment Fund (Grant RP03-064). JLM and BSM are supported by the National Health and Medical Research Council Career Development Fellowships (JLM, APP1066916; BSM, APP1004211). We thank Lance Macaulay and Sue Mitchell for critical reading and comments on the manuscript.

Author details

1CSIRO Food and Nutrition Flagship, PO Box 52, North Ryde, NSW 1670, Australia. 2CSIRO Agriculture Flagship, 306 Carmody Road, St Lucia, QLD 4067, Australia. 3Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia�4FOODplus Research Centre, Waite Campus, The University of Adelaide, PMB 1, Glen Osmond, SA 5064, Australia. 5Women�s and Children�s Health Research Institute, 72 King William Road, North Adelaide, SA 5006, Australia.

Blank
References:

1. WHO. WHO | Overweight and obesity. www.who.int/gho/ncd/
risk_factors/overweight/en/index.html. Accessed 29 January 2015.
2. Visscher PM, Brown MA, McCarthy MI, Yang J. Five years of GWAS discovery.
Am J Hum Genet. 2012;90:7�24.
3. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic
studies of body mass index yield new insights for obesity biology. Nature.
2015;518:197�206.
4. Ling C, Del Guerra S, Lupi R, R�nn T, Granhall C, Luthman H, et al.
Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and
effect on insulin secretion. Diabetologia. 2008;51:615�22.
5. Van Dijk SJ, Molloy PL, Varinli H, Morrison JL, Muhlhausler BS. Epigenetics
and human obesity. Int J Obes (Lond). 2015;39:85�97.
6. Teh AL, Pan H, Chen L, Ong M-L, Dogra S, Wong J, et al. The effect of
genotype and in utero environment on interindividual variation in neonate
DNA methylomes. Genome Res. 2014;24:1064�74.
7. Olsson AH, Volkov P, Bacos K, Dayeh T, Hall E, Nilsson EA, et al. Genomewide
associations between genetic and epigenetic variation influence
mRNA expression and insulin secretion in human pancreatic islets. PLoS
Genet. 2014;10:e1004735.
8. Grundberg E, Meduri E, Sandling JK, Hedman AK, Keildson S, Buil A, et al.
Global analysis of DNA methylation variation in adipose tissue from twins
reveals links to disease-associated variants in distal regulatory elements.
Am J Hum Genet. 2013;93:876�90.
9. Ronn T, Volkov P, Gillberg L, Kokosar M, Perfilyev A, Jacobsen AL, et al.
Impact of age, BMI and HbA1c levels on the genome-wide DNA
methylation and mRNA expression patterns in human adipose tissue
and identification of epigenetic biomarkers in blood. Hum Mol Genet.
2015;24:3792�813.
10. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental
origins hypothesis. Annu Rev Nutr. 2007;27:363�88.
11. McMillen IC, Rattanatray L, Duffield JA, Morrison JL, MacLaughlin SM, Gentili
S, et al. The early origins of later obesity: pathways and mechanisms. Adv
Exp Med Biol. 2009;646:71�81.
12. Ravelli A, van der Meulen J, Michels R, Osmond C, Barker D, Hales C, et al.
Glucose tolerance in adults after prenatal exposure to famine. Lancet.
1998;351:173�7.
13. McMillen IC, MacLaughlin SM, Muhlhausler BS, Gentili S, Duffield JL,
Morrison JL. Developmental origins of adult health and disease: the role of
periconceptional and foetal nutrition. Basic Clin Pharmacol Toxicol.
2008;102:82�9.
14. Zhang S, Rattanatray L, McMillen IC, Suter CM, Morrison JL. Periconceptional
nutrition and the early programming of a life of obesity or adversity. Prog
Biophys Mol Biol. 2011;106:307�14.
15. Bouret S, Levin BE, Ozanne SE. Gene-environment interactions controlling
energy and glucose homeostasis and the developmental origins of obesity.
Physiol Rev. 2015;95:47�82.
16. Borengasser SJ, Zhong Y, Kang P, Lindsey F, Ronis MJ, Badger TM, et al.
Maternal obesity enhances white adipose tissue differentiation and alters
genome-scale DNA methylation in male rat offspring. Endocrinology.
2013;154:4113�25.
17. Gluckman PD, Lillycrop KA, Vickers MH, Pleasants AB, Phillips ES, Beedle AS,
et al. Metabolic plasticity during mammalian development is directionally
dependent on early nutritional status. Proc Natl Acad Sci U S A.
2007;104:12796�800.
18. Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC, McLean C,
et al. Epigenetic gene promoter methylation at birth is associated with
child�s later adiposity. Diabetes. 2011;60:1528�34.
19. McMillen IC, Adam CL, Muhlhausler BS. Early origins of obesity:
programming the appetite regulatory system. J Physiol. 2005;565(Pt 1):9�17.
20. Begum G, Stevens A, Smith EB, Connor K, Challis JR, Bloomfield F, et al.
Epigenetic changes in fetal hypothalamic energy regulating pathways are
associated with maternal undernutrition and twinning. FASEB J.
2012;26:1694�703.
21. Ge ZJ, Liang QX, Hou Y, Han ZM, Schatten H, Sun QY, et al. Maternal obesity
and diabetes may cause DNA methylation alteration in the spermatozoa of
offspring in mice. Reprod Biol Endocrinol. 2014;12:29.
22. Jousse C, Parry L, Lambert-Langlais S, Maurin AC, Averous J, Bruhat A, et al.
Perinatal undernutrition affects the methylation and expression of the leptin
gene in adults: implication for the understanding of metabolic syndrome.
FASEB J. 2011;25:3271�8.
23. Lan X, Cretney EC, Kropp J, Khateeb K, Berg MA, Penagaricano F, et al.
Maternal diet during pregnancy induces gene expression and DNA
methylation changes in fetal tissues in sheep. Front Genet. 2013;4:49.
24. Li CC, Young PE, Maloney CA, Eaton SA, Cowley MJ, Buckland ME, et al.
Maternal obesity and diabetes induces latent metabolic defects and
widespread epigenetic changes in isogenic mice. Epigenetics. 2013;8:602�11.
25. Lillycrop KA, Phillips ES, Jackson AA, Hanson MA, Burdge GC. Dietary protein
restriction of pregnant rats induces and folic acid supplementation prevents
epigenetic modification of hepatic gene expression in the offspring. J Nutr.
2005;135:1382�6.
26. Radford EJ, Ito M, Shi H, Corish JA, Yamazawa K, Isganaitis E, et al. In utero
effects. In utero undernourishment perturbs the adult sperm methylome
and intergenerational metabolism. Science. 2014;345(80):1255903.
27. Suter M, Bocock P, Showalter L, Hu M, Shope C, McKnight R, et al.
Epigenomics: maternal high-fat diet exposure in utero disrupts
peripheral circadian gene expression in nonhuman primates. FASEB J.
2011;25:714�26.
28. Suter MA, Ma J, Vuguin PM, Hartil K, Fiallo A, Harris RA, et al. In utero
exposure to a maternal high-fat diet alters the epigenetic histone code in a
murine model. Am J Obs Gynecol. 2014;210:463 e1�463 e11.
29. Tosh DN, Fu Q, Callaway CW, McKnight RA, McMillen IC, Ross MG, et al.
Epigenetics of programmed obesity: alteration in IUGR rat hepatic IGF1
mRNA expression and histone structure in rapid vs. delayed postnatal
catch-up growth. Am J Physiol Gastrointest Liver Physiol.
2010;299:G1023�9.
30. Sandovici I, Smith NH, Nitert MD, Ackers-Johnson M, Uribe-Lewis S, Ito Y,
et al. Maternal diet and aging alter the epigenetic control of a promoterenhancer
interaction at the Hnf4a gene in rat pancreatic islets. Proc Natl
Acad Sci U S A. 2011;108:5449�54.
31. Braunschweig M, Jagannathan V, Gutzwiller A, Bee G. Investigations on
transgenerational epigenetic response down the male line in F2 pigs. PLoS
One. 2012;7, e30583.
32. Carone BR, Fauquier L, Habib N, Shea JM, Hart CE, Li R, et al. Paternally
induced transgenerational environmental reprogramming of metabolic
gene expression in mammals. Cell. 2010;143:1084�96.
33. Ost A, Lempradl A, Casas E, Weigert M, Tiko T, Deniz M, et al. Paternal diet
defines offspring chromatin state and intergenerational obesity. Cell.
2014;159:1352�64.
34. Mart�nez D, Pentinat T, Rib� S, Daviaud C, Bloks VW, Cebri� J, et al. In utero
undernutrition in male mice programs liver lipid metabolism in the secondgeneration
offspring involving altered Lxra DNA methylation. Cell Metab.
2014;19:941�51.
35. Wei Y, Yang C-R, Wei Y-P, Zhao Z-A, Hou Y, Schatten H, et al. Paternally
induced transgenerational inheritance of susceptibility to diabetes in
mammals. Proc Natl Acad Sci U S A. 2014;111:1873�8.
36. Grossniklaus U, Kelly WG, Kelly B, Ferguson-Smith AC, Pembrey M, Lindquist
S. Transgenerational epigenetic inheritance: how important is it? Nat Rev
Genet. 2013;14:228�35.
37. Pembrey M, Saffery R, Bygren LO. Human transgenerational responses to
early-life experience: potential impact on development, health and
biomedical research. J Med Genet. 2014;51:563�72.
38. Wolff GL, Kodell RL, Moore SR, Cooney CA. Maternal epigenetics and methyl
supplements affect agouti gene expression in Avy/a mice. FASEB J.
1998;12:949�57.
39. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility.
Nat Rev Genet. 2007;8:253�62.
40. Morgan HD, Sutherland HG, Martin DI, Whitelaw E. Epigenetic inheritance at
the agouti locus in the mouse. Nat Genet. 1999;23:314�8.
41. Cropley JE, Suter CM, Beckman KB, Martin DI. Germ-line epigenetic
modification of the murine A vy allele by nutritional supplementation. Proc
Natl Acad Sci U S A. 2006;103:17308�12.
42. Hoile SP, Lillycrop KA, Thomas NA, Hanson MA, Burdge GC. Dietary protein
restriction during F0 pregnancy in rats induces transgenerational changes in
the hepatic transcriptome in female offspring. PLoS One. 2011;6, e21668.
43. Multhaup ML, Seldin MM, Jaffe AE, Lei X, Kirchner H, Mondal P, et al. Mousehuman
experimental epigenetic analysis unmasks dietary targets and
genetic liability for diabetic phenotypes. Cell Metab. 2015;21:138�49.
44. Michels KB, Binder AM, Dedeurwaerder S, Epstein CB, Greally JM, Gut I, et al.
Recommendations for the design and analysis of epigenome-wide
association studies. Nat Methods. 2013;10:949�55.
45. Dayeh TA, Olsson AH, Volkov P, Almgren P, R�nn T, Ling C. Identification of
CpG-SNPs associated with type 2 diabetes and differential DNA methylation
in human pancreatic islets. Diabetologia. 2013;56:1036�46.
46. Relton CL, Davey Smith G. Two-step epigenetic Mendelian randomization: a
strategy for establishing the causal role of epigenetic processes in pathways
to disease. Int J Epidemiol. 2012;41:161�76.
47. Liu Y, Aryee MJ, Padyukov L, Fallin MD, Hesselberg E, Runarsson A, et al.
Epigenome-wide association data implicate DNA methylation as an
intermediary of genetic risk in rheumatoid arthritis. Nat Biotechnol.
2013;31:142�7.
48. Yuan W, Xia Y, Bell CG, Yet I, Ferreira T, Ward KJ, et al. An integrated
epigenomic analysis for type 2 diabetes susceptibility loci in monozygotic
twins. Nat Commun. 2014;5:5719.
49. Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E, et al. Impact of an
exercise intervention on DNA methylation in skeletal muscle from firstdegree
relatives of patients with type 2 diabetes. Diabetes. 2012;61:3322�32.
50. Gagnon F, A�ssi D, Carri� A, Morange P-E, Tr�gou�t D-A. Robust validation of
methylation levels association at CPT1A locus with lipid plasma levels.
J Lipid Res. 2014;55:1189�91.
51. Demerath EW, Guan W, Grove ML, Aslibekyan S, Mendelson M, Zhou Y-H,
et al. Epigenome-wide association atudy (EWAS) of BMI, BMI change, and
waist circumference in African American adults identifies multiple replicated
loci. Hum Mol Genet. 2015:ddv161�.
52. Dick KJ, Nelson CP, Tsaprouni L, Sandling JK, A�ssi D, Wahl S, et al. DNA
methylation and body-mass index: a genome-wide analysis. Lancet.
2014;6736:1�9.
53. Su S, Zhu H, Xu X, Wang X, Dong Y, Kapuku G, et al. DNA methylation of
the LY86 gene is associated with obesity, insulin resistance, and
inflammation. Twin Res Hum Genet. 2014;17:183�91.
54. Clarke-Harris R, Wilkin TJ, Hosking J, Pinkney J, Jeffery AN, Metcalf BS, et al.
PGC1? promoter methylation in blood at 5�7 years predicts adiposity from
9 to 14 years (EarlyBird 50). Diabetes. 2014;63:2528�37.
55. Guay S-P, Brisson D, Lamarche B, Biron S, Lescelleur O, Biertho L, et al.
ADRB3 gene promoter DNA methylation in blood and visceral adipose
tissue is associated with metabolic disturbances in men. Epigenomics.
2014;6:33�43.
56. Agha G, Houseman EA, Kelsey KT, Eaton CB, Buka SL, Loucks EB. Adiposity is
associated with DNA methylation profile in adipose tissue. Int J Epidemiol.
2014:1�11.
57. Irvin MR, Zhi D, Joehanes R, Mendelson M, Aslibekyan S, Claas SA, et al.
Epigenome-wide association study of fasting blood lipids in the genetics of
lipid-lowering drugs and diet network study. Circulation. 2014;130:565�72.
58. Frazier-Wood AC, Aslibekyan S, Absher DM, Hopkins PN, Sha J, Tsai MY, et al.
Methylation at CPT1A locus is associated with lipoprotein subfraction
profiles. J Lipid Res. 2014;55:1324�30.
59. Pfeifferm L, Wahl S, Pilling LC, Reischl E, Sandling JK, Kunze S, et al. DNA
methylation of lipid-related genes affects blood lipid levels. Circ Cardiovasc
Genet. 2015.
60. Petersen A-K, Zeilinger S, Kastenm�ller G, R�misch-Margl W, Brugger M, Peters
A, et al. Epigenetics meets metabolomics: an epigenome-wide association
study with blood serum metabolic traits. Hum Mol Genet. 2014;23:534�45.
61. Hidalgo B, Irvin MR, Sha J, Zhi D, Aslibekyan S, Absher D, et al. Epigenomewide
association study of fasting measures of glucose, insulin, and HOMA-IR
in the genetics of lipid lowering drugs and diet network study. Diabetes.
2014;63:801�7.
62. Dayeh T, Volkov P, Sal� S, Hall E, Nilsson E, Olsson AH, et al. Genome-wide
DNA methylation analysis of human pancreatic islets from type 2 diabetic
and non-diabetic donors identifies candidate genes that influence insulin
secretion. PLoS Genet. 2014;10, e1004160.
63. Nilsson E, Jansson PA, Perfilyev A, Volkov P, Pedersen M, Svensson MK, et al.
Altered DNA methylation and differential expression of genes influencing
metabolism and inflammation in adipose tissue from subjects with type 2
diabetes. Diabetes. 2014;63:2962�76.
64. Benton MC, Johnstone A, Eccles D, Harmon B, Hayes MT, Lea RA, et al. An analysis of DNA methylation in human adipose tissue reveals differential modification of obesity genes before and after gastric bypass and weight
loss. Gene. 2015;16:1�21.
65. Bateson P, Gluckman P. Plasticity and robustness in development and
evolution. Int J Epidemiol. 2012;41:219�23.
66. Feinberg AP, Irizarry RA, Feinberg AP, Irizarry RA. Evolution in health and
medicine Sackler colloquium: stochastic epigenetic variation as a driving
force of development, evolutionary adaptation, and disease. Proc Natl Acad
Sci U S A. 2010;107(Suppl):1757�64.
67. Martino D, Loke YJ, Gordon L, Ollikainen M, Cruickshank MN, Saffery R, et al.
Longitudinal, genome-scale analysis of DNA methylation in twins from birth
to 18 months of age reveals rapid epigenetic change in early life and pairspecific
effects of discordance. Genome Biol. 2013;14:R42.
68. Tobi EW, Goeman JJ, Monajemi R, Gu H, Putter H, Zhang Y, et al. DNA
methylation signatures link prenatal famine exposure to growth and
metabolism. Nat Commun. 2014;5:5592.
69. Dominguez-Salas P, Moore SE, Baker MS, Bergen AW, Cox SE, Dyer RA, et al.
Maternal nutrition at conception modulates DNA methylation of human
metastable epialleles. Nat Commun. 2014;5:3746.
70. Quilter CR, Cooper WN, Cliffe KM, Skinner BM, Prentice PM, Nelson L, et al.
Impact on offspring methylation patterns of maternal gestational diabetes
mellitus and intrauterine growth restraint suggest common genes and
pathways linked to subsequent type 2 diabetes risk. FASEB J. 2014:1�12.
71. Morales E, Groom A, Lawlor DA, Relton CL. DNA methylation signatures in
cord blood associated with maternal gestational weight gain: results from
the ALSPAC cohort. BMC Res Notes. 2014;7:278.
72. Ruchat SM, Houde AA, Voisin G, St-Pierre J, Perron P, Baillargeon JP, et al.
Gestational diabetes mellitus epigenetically affects genes predominantly
involved in metabolic diseases. Epigenetics. 2013;8:935�43.
73. Liu X, Chen Q, Tsai H-J, Wang G, Hong X, Zhou Y, et al. Maternal
preconception body mass index and offspring cord blood DNA
methylation: exploration of early life origins of disease. Environ Mol
Mutagen. 2014;55:223�30.
74. Soubry A, Murphy SK, Wang F, Huang Z, Vidal AC, Fuemmeler BF, et al.
Newborns of obese parents have altered DNA methylation patterns at
imprinted genes. Int J Obes (Lond). 2015;39:650�7.
75. Jacobsen SC, Br�ns C, Bork-Jensen J, Ribel-Madsen R, Yang B, Lara E, et al.
Effects of short-term high-fat overfeeding on genome-wide DNA
methylation in the skeletal muscle of healthy young men. Diabetologia.
2012;55:3341�9.
76. Gillberg L, Jacobsen SC, R�nn T, Br�ns C, Vaag A. PPARGC1A DNA
methylation in subcutaneous adipose tissue in low birth weight subjects�
impact of 5 days of high-fat overfeeding. Metabolism. 2014;63:263�71.
77. Huang Y-T, Maccani JZJ, Hawley NL, Wing RR, Kelsey KT, McCaffery JM.
Epigenetic patterns in successful weight loss maintainers: a pilot study. Int J
Obes (Lond). 2015;39:865�8.
78. Barres R, Kirchner H, Rasmussen M, Yan J, Kantor FR, Krook A, N�slund E,
Zierath JR. Weight loss after gastric bypass surgery in human obesity
remodels promoter methylation. Cell Rep. 2013:1�8.
79. Ahrens M, Ammerpohl O, von Sch�nfels W, Kolarova J, Bens S, Itzel T, et al.
DNA methylation analysis in nonalcoholic fatty liver disease suggests
distinct disease-specific and remodeling signatures after bariatric surgery.
Cell Metab. 2013;18:296�302.
80. Voisin S, Eynon N, Yan X, Bishop DJ. Exercise training and DNA methylation
in humans. Acta Physiol (Oxf). 2014;213:39�59.
81. Lindholm ME, Marabita F, Gomez-Cabrero D, Rundqvist H, Ekstr�m TJ,
Tegn�r J, et al. An integrative analysis reveals coordinated reprogramming
of the epigenome and the transcriptome in human skeletal muscle after
training. Epigenetics. 2014;9:1557�69.
82. Denham J, O�Brien BJ, Marques FZ, Charchar FJ. Changes in the leukocyte
methylome and its effect on cardiovascular related genes after exercise.
J Appl Physiol. 2014:jap.00878.2014.
83. Rowlands DS, Page RA, Sukala WR, Giri M, Ghimbovschi SD, Hayat I, et al.
Multi-omic integrated networks connect DNA methylation and miRNA with
skeletal muscle plasticity to chronic exercise in type 2 diabetic obesity.
Physiol Genomics. 2014;46:747�65.
84. Horvath S, Erhart W, Brosch M, Ammerpohl O, von Schonfels W, Ahrens M,
et al. Obesity accelerates epigenetic aging of human liver. Proc Natl Acad
Sci. 2014;111:15538�43.
85. Alm�n MS, Nilsson EK, Jacobsson JA, Kalnina I, Klovins J, Fredriksson R, et al.
Genome-wide analysis reveals DNA methylation markers that vary with
both age and obesity. Gene. 2014.;548:61�7
86. Houseman EA, Molitor J, Marsit CJ. Reference-free cell mixture adjustments
in analysis of DNA methylation data. Bioinformatics. 2014;30:1431�9.
87. Wells JC. A critical appraisal of the predictive adaptive response hypothesis.
Int J Epidemiol. 2012;41:229�35.
88. Williams-Wyss O, Zhang S, MacLaughlin SM, Kleemann D, Walker SK, Suter
CM, et al. Embryo number and periconceptional undernutrition in the
sheep have differential effects on adrenal epigenotype, growth, and
development. Am J Physiol Endocrinol Metab. 2014;307:E141�50.
89. Zhang S, Rattanatray L, Morrison JL, Nicholas LM, Lie S, McMillen IC.
Maternal obesity and the early origins of childhood obesity: weighing up
the benefits and costs of maternal weight loss in the periconceptional
period for the offspring. Exp Diabetes Res. 2011;2011:585749.
90. Zhang S, Williams-Wyss O, MacLaughlin SM, Walker SK, Kleemann DO, Suter
CM, et al. Maternal undernutrition during the first week after conception
results in decreased expression of glucocorticoid receptor mRNA in the
absence of GR exon 17 hypermethylation in the fetal pituitary in late
gestation. J Dev Orig Heal Dis. 2013;4:391�401.
91. Lie S, Morrison JL, Williams-Wyss O, Suter CM, Humphreys DT, Ozanne SE,
et al. Periconceptional undernutrition programs changes in insulin-signaling
molecules and microRNAs in skeletal muscle in singleton and twin fetal
sheep. Biol Reprod. 2014;90:5.
92. Van Straten EM, van Meer H, Huijkman NC, van Dijk TH, Baller JF, Verkade
HJ, et al. Fetal liver X receptor activation acutely induces lipogenesis but
does not affect plasma lipid response to a high-fat diet in adult mice. Am J
Physiol Endocrinol Metab. 2009;297:E1171�8.
93. Fernandez-Twinn DS, Alfaradhi MZ, Martin-Gronert MS, Duque-Guimaraes
DE, Piekarz A, Ferland-McCollough D, et al. Downregulation of IRS-1 in
adipose tissue of offspring of obese mice is programmed cellautonomously
through post-transcriptional mechanisms. Mol Metab.
2014;3:325�33.
94. Waterland RA, Travisano M, Tahiliani KG. Diet-induced hypermethylation at
agouti viable yellow is not inherited transgenerationally through the female.
FASEB J. 2007;21:3380�5.
95. Ge ZJ, Luo SM, Lin F, Liang QX, Huang L, Wei YC, et al. DNA methylation in
oocytes and liver of female mice and their offspring: effects of high-fat-dietinduced
obesity. Env Heal Perspect. 2014;122:159�64.
96. Ollikainen M, Ismail K, Gervin K, Kyll�nen A, Hakkarainen A, Lundbom J, et al.
Genome-wide blood DNA methylation alterations at regulatory elements
and heterochromatic regions in monozygotic twins discordant for obesity
and liver fat. Clin Epigenetics. 2015;7:1�13.

Close Accordion
Body Composition Evaluation: A Clinical Practice Tool

Body Composition Evaluation: A Clinical Practice Tool

Body Composition: Key Words

  • Fat-free mass
  • Fat mass
  • Undernutrition
  • Bioelectrical impedance analysis
  • Sarcopenic obesity
  • Drug toxicity

Abstract

Undernutrition is insufficiently detected in in- and outpatients, and this is likely to worsen during the next decades. The increased prevalence of obesity together with chronic illnesses associated with fat-free mass (FFM) loss will result in an increased prevalence of sarcopenic obesity. In patients with sarcopenic obesity, weight loss and the body mass index lack accuracy to detect FFM loss. FFM loss is related to increasing mortality, worse clinical outcomes, and impaired quality of life. In sarcopenic obesity and chronic diseases, body composition measurement with dual-energy X-ray absorptiometry, bioelectrical impedance analysis, or computerized tomography quantifies the loss of FFM. It allows tailored nutritional support and disease-specific therapy and reduces the risk of drug toxicity. Body composition evaluation should be integrated into routine clinical practice for the initial assessment and sequential follow-up of nutritional status. It could allow objective, systematic, and early screening of undernutrition and promote the rational and early initiation of optimal nutritional support, thereby contributing to reducing malnutrition-induced morbidity, mortality, worsening of the quality of life, and global health care costs.

Introduction

man overweight 3D modelChronic undernutrition is characterized by a progressive reduction of the�fat-free mass (FFM) and fat mass (FM)�and �which has deleterious consequences on health. Undernutrition is insufficiently screened and treated in hospitalized or at-risk patients despite its high prevalence and negative impact on mortality, morbidity, length of stay (LOS), quality of life, and costs [1�4]. The risk of underestimating hospital undernutrition is likely to worsen in the next decades because of the increasing prevalence of overweight, obesity, and chronic diseases and the increased number of elderly subjects. These clinical conditions are associated with FFM loss (sarcopenia). Therefore, an increased number of patients with FFM loss and sarcopenic obesity will be seen in the future.

Sarcopenic obesity is associated with decreased survival and increased therapy toxicity in cancer patients [5�10], whereas FFM loss is related to decreased survival, a negative clinical outcome, increased health care costs [2], and impaired overall health, functional capacities, and quality of life [4�11]. Therefore, the detection and treatment of FFM loss is a major issue of public health and health costs [12].

Weight loss and the body mass index (BMI) lack sensitivity to detect FFM loss [13]. In this review, we support the systematic assessment of FFM with a method of body composition evaluation in order to improve the detection, management, and follow-up of undernutrition. Such an approach should in turn reduce the clinical and functional consequences of diseases in the setting of a cost- effective medico-economic approach (fig. 1). We discuss the main applications of body composition evaluation in clinical practice (fig. 2).

body composition fig 1

Fig. 1. Conceptualization of the expected impact of early use of body composition for the screening of fat-free loss and�under-nutrition in sarcopenic overweight and obese subjects. An increased prevalence of overweight and obesity is observed in all Western and emerging countries. Simultaneously, the aging of the population, the reduction of the level of physical activity, and the higher prevalence of chronic dis- eases and cancer increased the number of patients with or at risk of FFM impairment, i.e. sarcopenia. Thus, more patients are presenting with �sarcopenic over- weight or obesity�. In these patients, evaluation of nutritional status using anthropometric methods, i.e. weight loss and calculation of BMI, is not sensitive enough to detect FFM impairment. As a result, undernutrition is not detected, worsens, and negatively impacts morbidity, mortality, LOS, length of recovery, quality of life, and health care costs. On the contrary, in patients with �sarcopenic overweight or obesity�, early screening of undernutrition with a dedicated method of body composition evaluation would allow early initiation of nutritional support and, in turn, improvements of nutritional status and clinical outcome.

Rationale for a New Strategy for the Screening of Undernutrition

Screening of Undernutrition Is Insufficient

checklistAcademic societies encourage systematic screening of undernutrition at hospital admission and during the hospital stay [14]. The detection of undernutrition is generally based on measurements of weight and height, calculations of BMI, and the percentage of weight loss. Nevertheless, screening of undernutrition is infrequent in hospitalized or nutritionally at-risk ambulatory patients. For example, in France, surveys performed by the French Health Authority [15] indicate that: (i) weight alone, (ii) weight with BMI or percentage of weight loss, and (iii) weight, BMI,�and percentage of weight loss are reported in only 55, 30, and 8% of the hospitalized patients� records, respectively. Several issues, which could be improved by specific educational programs, explain the lack of implementation of nutritional screening in hospitals (table 1). In addition, the accuracy of the clinical screening of undernutrition could be limited at hospital admission. Indeed, patients with undernutrition may have the same BMI as sex- and age- matched healthy controls but a significantly decreased FFM hidden by an expansion of the FM and the total body water which can be measured by bioelectrical impedance analysis (BIA) [13]. This example illustrates that body composition evaluation allows a more accurate identification of FFM loss than body weight loss or BMI decrease. The lack of sensitivity and specificity of weight, BMI, and percentage of weight loss argue for the need for other methods to evaluate the nutritional status.

Changes in Patients� Profiles

patient consulting a doctorIn 2008, twelve and thirty percent of the worldwide adult population was obese or overweight; this is two times higher than in 1980 [16]. The prevalence of overweight and obesity is also increasing in hospitalized patients. A 10-year comparative survey performed in a European hospital showed an increase in patients� BMI, together with a shorter LOS [17]. The BMI increase masks undernutrition and FFM loss at hospital admission. The increased prevalence of obesity in an aging population has led to the recognition of a new nutritional entity: �sarcopenic obesity� [18]. Sarcopenic obesity is characterized by increased FM and reduced FFM with a normal or high body weight. The emergence of the concept of sarcopenic obesity will increase the number of situations associated with a lack of sensitivity of the calculations of BMI and�body weight change for the early detection of FFM loss. This supports a larger use of body composition evaluation for the assessment and follow-up of nutritional status in clinical practice (fig. 1).

body composition fig 2Fig. 2. Current and potential applications of body composition evaluation in clinical practice. The applications are indicated in the boxes, and the body composition methods that could be used for each application are indicated inside the circles. The most used application of body composition evaluation is the measurement of bone mineral density by DEXA for the diagnosis and management of osteoporosis. Although a low FFM is associated with worse clinical outcomes, FFM evaluation is not yet implemented enough in clinical practice. However, by allowing early detection of undernutrition, body composition evaluation could improve the clinical outcome. Body composition evaluation could also be used to follow up nutritional status, calculate energy needs, tailor nutritional support, and assess fluid changes during perioperative period and renal insufficiency. Recent evidence indicates that�a low FFM is associated with a higher toxicity of some chemo- therapy drugs in cancer patients. Thus, by allowing tailoring of the chemotherapy doses to the FFM in cancer patients, body com- position evaluation should improve the tolerance and the efficacy of chemotherapy. BIA, L3-targeted CT, and DEXA could be used for the assessment of nutritional status, the calculation of energy needs, and the tailoring of nutritional support and therapy. Further studies are warranted to validate BIA as an accurate method for fluid balance measurement. By integrating body composition evaluation into the management of different clinical conditions, all of these potential applications would lead to a better recognition of nutritional care by the medical community, the health care facilities, and the health authorities, as well as to an increase in the medico-economic benefits of the nutritional evaluation.

Body Composition Evaluation For The Assessment Of Nutritional Status

Body composition evaluation is a valuable technique to assess nutritional status. Firstly, it gives an evaluation of nutritional status through the assessment of FFM. Secondly, by measuring FFM and phase angle with BIA, it allows evaluation of the disease prognosis and outcome.

body composition table 1

body composition table 2Body Composition Techniques For FFM Measurement

Body composition evaluation allows measurement of the major body compartments: FFM (including bone mineral tissue), FM, and total body water. Table 2 shows indicative values of the body composition of a healthy subject weighing 70 kg. In several clinical situations, i.e. hospital admission, chronic obstructive pulmonary dis- ease (COPD) [21�23], dialysis [24�26], chronic heart failure [27], amyotrophic lateral sclerosis [28], cancer [5, 29], liver transplantation [30], nursing home residence [31], and Alzheimer�s disease [32], changes in body compartments are detected with the techniques of body composition evaluation. At hospital admission, body composition evaluation could be used for the detection of FFM loss and undernutrition. Indeed, FFM and the FFM index (FFMI) [FFM (kg)/height (m2)] measured by BIA are significantly lower in hospitalized patients (n = 995) than in age-, height-, and sex-matched controls (n = 995) [3]. Conversely, clinical tools of nutritional status assessment, such as BMI, subjective global assessment, or mini-nutritional assessment, are not accurate enough to estimate FFM loss and nutritional status [30, 32�34]. In 441 patients with non-small cell lung cancer, FFM loss deter- mined by computerized tomography (CT) was observed in each BMI category [7], and in young adults with all�types of cancer, an increase in FM together with a de- crease in FFM were reported [29]. These findings reveal the lack of sensitivity of BMI to detect FFM loss. More- over, the FFMI is a more sensitive determinant of LOS than a weight loss over 10% or a BMI below 20 [3]. In COPD, the assessment of FFM by BIA is a more sensitive method to detect undernutrition than anthropometry [33, 35]. BIA is also more accurate at assessing nutrition- al status in children with severe neurologic impairment than the measurement of skin fold thickness [36].

Body Composition For The Evaluation Of Prognosis & Clinical Outcome

FFM loss is correlated with survival in different clinical settings [5, 21�28, 37]. In patients with amyotrophic lateral sclerosis, an FM increase, but not an FFM in- crease, measured by BIA, was correlated with survival during the course of the disease [28]. The relation between body composition and mortality has not yet been demonstrated in the intensive care unit. The relation between body composition and mortality has been demonstrated with anthropometric methods, BIA, and CT. Measurement of the mid-arm muscle circumference is an easy tool to diagnose sarcopenia [38]. The mid-arm muscle circumference has been shown to be correlated with survival in patients with cirrhosis [39, 40], HIV infection [41], and COPD in a stronger way than BMI [42]. The relation between FFM loss and mortality has been extensively shown with BIA [21�28, 31, 37], which is the most used method. Recently, very interesting data suggest that CT could evaluate the disease prognosis in relation to muscle wasting. In obese cancer patients, sarcopenia as assessed by CT measurement of the total skeletal muscle cross-sectional area is an independent predictor of the survival of patients with bronchopulmonary [5, 7], gastrointestinal [5], and pancreatic cancers [6]. FFM assessed by measurement of the mid-thigh muscle cross- sectional area by CT is also predictive of mortality in COPD patients with severe chronic respiratory insufficiency [43]. In addition to mortality, a low FFMI at hospital admission is significantly associated with an in- creased LOS [3, 44]. A bicentric controlled population study performed in 1,717 hospitalized patients indicates that both loss of FFM and excess of FM negatively affect the LOS [44]. Patients with sarcopenic obesity are most at risk of increased LOS. This study also found that ex- cess FM reduces the sensitivity of BMI to detect nutritional depletion [44]. Together with the observation that the BMI of hospitalized patients has increased during the last decade [17], these findings suggest that FFM and�FFMI measurement should be used to evaluate nutritional status in hospitalized patients.

BIA measures the phase angle [45]. A low phase angle is related to survival in oncology [46�50], HIV infection/ AIDS [51], amyotrophic lateral sclerosis [52], geriatrics [53], peritoneal dialysis [54], and cirrhosis [55]. The phase angle threshold associated with reduced survival is variable: less than 2.5 degrees in amyotrophic lateral sclerosis patients [52], 3.5 degrees in geriatric patients [53], from less than 1.65 to 5.6 degrees in oncology patients [47�50], and 5.4 degrees in cirrhotic patients [55]. The phase angle is also associated with the severity of lymphopenia in AIDS [56], and with the risk of postoperative complications among gastrointestinal surgical patients [57]. The relation of phase angle with prognosis and disease severity reinforces the interest in using BIA for the clinical management of patients with chronic diseases at high risk of undernutrition and FFM loss.

In summary, FFM loss or a low phase angle is related to mortality in patients with chronic diseases, cancer (in- cluding obesity cancer patients), and elderly patients in long-stay facilities. A low FFM and an increased FM are associated with an increased LOS in adult hospitalized patients. The relation between FFM loss and clinical out- come is clearly shown in patients with sarcopenic obesity. In these patients, as the sensitivity of BMI for detecting FFM loss is strongly reduced, body composition evalua- tion appears to be the method of choice to detect under- nutrition in routine practice. Overall, the association between body composition, phase angle, and clinical outcome reinforces the pertinence of using a body com- position evaluation in clinical practice.

Which Technique Of Body Composition Evaluation Should Be Used For The Assessment Of Nutritional Status?

Numerous methods of body composition evaluation have been developed: anthropometry, including the 4-skinfold method [58], hydrodensitometry [58], in vivo neutron activation analysis [59], anthropogammametry from total body potassium-40 [60], nuclear magnetic resonance [61], dual-energy X-ray absorptiometry (DEXA) [62, 63], BIA [45, 64�66], and more recently CT [7, 43, 67]. DEXA, BIA, and CT appear to be the most convenient methods for clinical practice (fig. 2), while the other methods are reserved for scientific use.

Compared with other techniques of body composition evaluation, the lack of reproducibility and sensitivity of the 4-skinfold method limits its use for the accurate measurement of body composition in clinical practice [33,�34]. However, in patients with cirrhosis [39, 40], COPD [34], and HIV infection [41], measurement of the mid- arm muscle circumference could be used to assess sarcopenia and disease-related prognosis. DEXA allows non- invasive direct measurement of the three major components of body composition. The measurement of bone mineral tissue by DEXA is used in clinical practice for the diagnosis and follow-up of osteoporosis. As the clinical conditions complicated by osteoporosis are often associated with undernutrition, i.e. elderly women, patients with organ insufficiencies, COPD [68], inflammatory bowel diseases, and celiac disease, DEXA could be of the utmost interest for the follow-up of both osteoporosis and nutritional status. However, the combined evaluation of bone mineral density and nutritional status is difficult to implement in clinical practice because the reduced accessibility of DEXA makes it impossible to be performed in all nutritionally at-risk or malnourished patients. The principles and clinical utilization of BIA have been largely described in two ESPEN position papers [45, 66]. BIA is based on the capacity of hydrated tissues to conduct electrical energy. The measurement of total body impedance allows estimation of total body water by assuming that total body water is constant. From total body water, validated equations allow the calculation of FFM and FM [69], which are interpreted according to reference values [70]. BIA is the only technique which allows calculation of the phase angle, which is correlated with the prognosis of various diseases. BIA equations are valid for: COPD [65]; AIDS wasting [71]; heart, lung, and liver transplantation [72]; anorexia nervosa [73] patients, and elderly subjects [74]. However, no BIA-specific equations have been validated in patients with extreme BMI (less than 17 and higher than 33.8) and dehydration or fluid overload [45, 66]. Nevertheless, because of its simplicity, low cost, quickness of use at bedside, and high interoperator reproducibility, BIA appears to be the technique of choice for the systematic and repeated evaluation of FFM in clinical practice, particularly at hospital admission and in chronic diseases. Finally, through written and objective re- ports, the wider use of BIA should allow improvement of the traceability of nutritional evaluation and an increase in the recognition of nutritional care by the health authorities. Recently, several data have suggested that CT images targeted on the 3rd lumbar vertebra (L3) could strongly predict whole-body fat and FFM in cancer patients, as compared with DEXA [7, 67]. Interestingly, the evaluation of body composition by CT presents great practical significance due to its routine use in patient diagnosis, staging, and follow-up. L3-targeted CT images�evaluate FFM by measuring the muscle cross-sectional area from L3 to the iliac crest by use of Hounsfield unit (HU) thresholds (�29 to +150) [5, 7]. The muscles included in the calculation of the muscle cross-sectional area are psoas, paraspinal muscles (erector spinae, quadratus lumborum), and abdominal wall muscles (transversus abdominis, external and internal obliques, rectus ab- dominis) [6]. CT also provided detail on specific muscles, adipose tissues, and organs not provided by DEXA or BIA. L3-targeted CT images could be theoretically per- formed solely, since they result in X-ray exposition similar to that of a chest radiography.

In summary, DEXA, BIA, and L3-targeted CT images could all measure body composition accurately. The technique selection will depend on the clinical context, hard- ware, and knowledge availability. Body composition evaluation by DEXA should be performed in patients having a routine assessment of bone mineral density. Also, analysis of L3-targeted CT is the method of choice for body composition evaluation in cancer patients. Body composition evaluation should also be done for every abdominal CT performed in patients who are nutritionally at risk or undernourished. Because of its simplicity of use, BIA could be widely implemented as a method of body com- position evaluation and follow-up in a great number of hospitalized and ambulatory patients. Future research will aim to determine whether a routine evaluation of body composition would allow early detection of the in- creased FFM catabolism related to critical illness [75].

Body Composition Evaluation For The Calculation Of Energy Needs

vegetable-juicesThe evaluation of FFM could be used for the calculation of energy needs, thus allowing the optimization of nutritional intakes according to nutritional needs. This could be of great interest in specific situations, such as severe neurologic disability, overweight, and obesity. In 61 children with severe neurologic impairment and intellectual disability, an equation integrating body composition had good agreement with the doubly labeled water method. It gave a better estimation of energy expenditure than did the Schofield predictive equation [36]. However, in 9 anorexia nervosa patients with a mean BMI of 13.7, pre- diction formulas of resting energy expenditure including FFM did not allow accurate prediction of the resting energy expenditure measured by indirect calorimetry [76]. In overweight or obese patients, the muscle catabolism in response to inflammation was the same as that observed�in patients with normal BMI. Indeed, despite a higher BMI, the FFM of overweight or obese individuals is similar (or slightly increased) to that of patients with normal BMI. Thus, the use of actual weight for the assessment of the energy needs of obese patients would result in over- feeding and its related complications. Therefore, the ex- perts recommend the use of indirect calorimetry or calculation of the energy needs of overweight or obese patients as follows: 15 kcal/kg actual weight/day or 20�25 kcal/kg ideal weight/day [77, 78], although these predictive formulas could be inaccurate in some clinical conditions [79]. In a US prospective study conducted in 33 ICU medical and surgical ventilated ICU patients, daily measurement of the active cell mass (table 2) by BIA was used to assess the adequacy between energy/protein intakes and needs. In that study, nutritional support with 30 kcal/ kg actual body weight/day energy and 1.5 g/kg/day protein allowed stabilization of the active cell mass [75]. Thus, follow-up of FFM by BIA could help optimize nutritional intakes when indirect calorimetry cannot be performed.

In summary, the measurement of FFM should help ad- just the calculation of energy needs (expressed as kcal/kg FFM) and optimize nutritional support in critical cases other than anorexia nervosa.

Body Composition Evaluation For The Follow-Up & Tailoring Of Nutritional Support

towel different nutritionBody composition evaluation allows a qualitative assessment of body weight variations. The evaluation of body composition may help to document the efficiency of nutritional support during a patient�s follow-up of numerous clinical conditions, such as surgery [59], anorexia nervosa [76, 80], hematopoietic stem cell transplantation [81], COPD [82], ICU [83], lung transplantation [84], ulcerative colitis [59], Crohn�s disease [85], cancer [86, 87], HIV/AIDS [88], and acute stroke in elderly patients [89]. Body composition evaluation could be used for the follow-up of healthy elderly subjects [90]. Body composition evaluation allows characterization of the increase in body mass in terms of FFM and FM [81, 91]. After hematopoietic stem cell transplantation, the increase in BMI is the result of the increase in FM, but not of the increase in FFM [81]. Also, during recovery after an acute illness, weight gain 6 months after ICU discharge could be mostly related to an increase in FM (+7 kg) while FFM only increased by 2 kg; DEXA and air displacement plethysmography were used to measure the FM and FFM [91]. These two examples suggest that body composition evaluation could be helpful to decide the modification and/or the renewal of nutritional support. By identifying the patients gaining weight but reporting no or insufficient FFM, body composition evaluation could contribute to influencing the medical decision of continuing nutrition- al support that would have been stopped in the absence of body composition evaluation.

In summary, body composition evaluation is of the utmost interest for the follow-up of nutritional support and its impact on body compartments.

Body Composition Evaluation For Tailoring Medical Treatments

In clinical situations when weight and BMI do not reflect the FFM, the evaluation of body composition should be used to adapt drug doses to the FFM and/or FM absolute values in every patient. This point has been recently illustrated in oncology patients with sarcopenic obesity. FFM loss was determined by CT as described above. In cancer patients, some therapies could affect body com- position by inducing muscle wasting [92]. In patients with advanced renal cell carcinoma [92], sorafenib induces a significant 8% loss of skeletal muscular mass at 12 months. In turn, muscle wasting in patients with BMI less than 25 was significantly associated with sorafenib toxicity in patients with metastatic renal cancer [8]. In metastatic breast cancer patients receiving capecitabine treatment, and in patients with colorectal cancer receiving 5-fluorouracile, using the convention of dosing per unit of body surface area, FFM loss was the determinant of chemotherapy toxicity [9, 10] and time to tumor progression [10]. In colorectal cancer patients administered 5-fluoruracil, low FFM is a significant predictor of toxicity only in female patients [9]. The variation in toxicity between women and men may be partially explained by the fact that FFM was lower in females. Indeed, FFM rep- resents the distribution volume of most cytotoxic chemo- therapy drugs. In 2,115 cancer patients, the individual variations in FFM could change by up to three times the distribution volume of the chemotherapy drug per body area unit [5]. Thus, administering the same doses of chemotherapy drugs to a patient with a low FFM compared to a patient with a normal FFM would increase the risk of chemotherapy toxicity [5]. These data suggest that FFM loss could have a direct impact on the clinical outcome of cancer patients. Decreasing chemotherapy doses in case of FFM loss could contribute to improving cancer patients� prognosis through the improvement of the tolerance of chemotherapy. These findings justify the systematic evaluation of body composition in all cancer patients in order to detect FFM loss, tailor chemotherapy doses according to FFM values, and then improve the efficacy- tolerance and cost-efficiency ratios of the therapeutic strategies [93]. Body composition evaluation should also be used to tailor the doses of drugs which are calculated based on patients� weight, e.g. corticosteroids, immuno-suppressors (infliximab, azathioprine or methotrexate), or sedatives (propofol).

In summary, measurement of FFM should be implemented in cancer patients treated with chemotherapy. Clinical studies are needed to demonstrate the importance of measuring body composition in patients treated with other medical treatments.

Towards The Implementation Of Body Composition Evaluation In Clinical Practice

When There's No Cure For Your Aching Back E-book Cover

News Letter

hypertension blood pressure pillsThe implementation of body composition evaluation in routine care presents a challenge for the next decades. Indeed the concomitant increases in elderly subjects and patients with chronic diseases and cancer, and in the prevalence of overweight and obesity in the population, will increase the number of patients nutritionally at risk or undernourished, particularly those with sarcopenic obesity. Body composition evaluation should be used to improve the screening of undernutrition in hospitalized patients. The results of body composition should be based on the same principle as BMI calculation, towards the systematic normalization for body height of FFM (FFMI) and FM [FM (kg)/height (m)2 = FM index] [94]. The results could be expressed according to previously de- scribed percentiles of healthy subjects [95, 96]. Body com- position evaluation should be performed at the different stages of the disease, during the course of treatments and the rehabilitation phase. Such repeated evaluations of body composition could allow assessment of the nutritional status, adjusting the calculation of energy needs as kilocalories/kilogram FFM, following the efficacy of nutritional support, and tailoring drug and nutritional therapies. BIA, L3-targeted CT, and DEXA represent the techniques of choice to evaluate body composition in clinical practice (fig. 2). In the setting of cost-effective and pragmatic use, these three techniques should be alternatively chosen. In cancer, undernourished, and nutritionally at-risk patients, an abdominal CT should be completed by the analysis of L3-targeted images for the evaluation of body composition.

In other situations, BIA appears to be the simplest most reproducible and less expensive method, while DEXA, if feasible, remains the reference method for clinical practice. By allowing earlier management of undernutrition, body composition evaluation can contribute to reducing malnutrition-induced morbidity and mortality, improving the quality of life and, as a consequence, increasing the medico-economic benefits (fig. 1). The latter needs to be demonstrated. Moreover, based on a more scientific approach, i.e. allowing for printing reports, objective initial assessment and follow-up of nutritional status, and the adjustment of drug doses, body composition evaluation would contribute to a better recognition of the activities related to nutritional evaluation and care by the medical community, health care facilities, and health authorities (fig. 2).

Conclusion

woman buying fresh organic vegetables

Screening of undernutrition is insufficient to allow for optimal nutrition care. This is in part due to the lack of sensitivity of BMI and weight loss for detecting FFM loss in patients with chronic diseases. Methods of body com- position evaluation allow a quantitative measurement of FFM changes during the course of disease and could be used to detect FFM loss in the setting of an objective, systematic, and early undernutrition screening. FFM loss is closely related to impaired clinical outcomes, survival, and quality of life, as well as increased therapy toxicity in cancer patients. Thus, body composition evaluation should be integrated into clinical practice for the initial assessment, sequential follow-up of nutritional status, and the tailoring of nutritional and disease-specific therapies. Body composition evaluation could contribute to strengthening the role and credibility of nutrition in the global medical management, reducing the negative impact of malnutrition on the clinical outcome and quality of life, thereby increasing the overall medico-economic benefits.

Acknowledgements

R. Thibault and C. Pichard are supported by research grants from the public foundation Nutrition 2000 Plus.

Disclosure Statement

Ronan Thibault and Claude Pichard declare no conflict of interest.

 

blank
References:

1 Pirlich M, Schutz T, Norman K, Gastell S,
L�bke HJ, Bischoff SC, Bolder U, Frieling
T, G�ldenzoph H, Hahn K, Jauch KW,
Schindler K, Stein J, Volkert D, Weimann A,
Werner H, Wolf C, Z�rcher G, Bauer P, Lochs
H: The German hospital malnutrition study.
Clin Nutr 2006;25:563�572.
2 Amaral TF, Matos LC, Tavares MM, Subtil
A, Martins R, Nazar� M, Sousa Pereira N:
The economic impact of disease-related malnutrition
at hospital admission. Clin Nutr
2007;26:778�784.
3 Pichard C, Kyle UG, Morabia A, Perrier A,
Vermeulen B, Unger P: Nutritional assessment:
lean body mass depletion at hospital
admission is associated with increased
length of stay. Am J Clin Nutr 2004;79:613�
618.
4 Capuano G, Gentile PC, Bianciardi F, Tosti
M, Palladino A, Di Palma M: Prevalence and
influence of malnutrition on quality of life
and performance status in patients with locally
advanced head and neck cancer before
treatment. Support Care Cancer 2010;18:
433�437.
5 Prado CM, Lieffers JR, McCargar LJ, Reiman
T, Sawyer MB, Martin L, Baracos VE: Prevalence
and clinical implications of sarcopenic
obesity in patients with solid tumours of the
respiratory and gastrointestinal tracts: a
population-based study. Lancet Oncol 2008;
9:629�635.
6 Tan BHL, Birdsell LA, Martin L, Baracos VE,
Fearon KC: Sarcopenia in an overweight or
obese patient is an adverse prognostic factor
in pancreatic cancer. Clin Cancer Res 2009;
15:6973�6979.
7 Baracos VE, Reiman T, Mourtzakis M,
Gioulbasanis I, Antoun S: Body composition
in patients with non-small cell lung cancer:
a contemporary view of cancer cachexia
with the use of computed tomography image
analysis. Am J Clin Nutr 2010;91(suppl):
1133S�1137S.
8 Antoun S, Baracos VE, Birdsell L, Escudier
B, Sawyer MB: Low body mass index and sarcopenia
associated with dose-limiting toxicity
of sorafenib in patients with renal cell carcinoma.
Ann Oncol 2010;21:1594�1598
9 Prado CM, Baracos VE, McCargar LJ,
Mourtzakis M, Mulder KE, Reiman T, Butts
CA, Scarfe AG, Sawyer MB: Body composition
as an independent determinant of 5-fluorouracil-based
chemotherapy toxicity. Clin
Cancer Res 2007;13:3264�3268.
10 Prado CM, Baracos VE, McCargar LJ, Reiman
T, Mourtzakis M, Tonkin K, Mackey JR,
Koski S, Pituskin E, Sawyer MB: Sarcopenia
as a determinant of chemotherapy toxicity
and time to tumor progression in metastatic
breast cancer patients receiving capecitabine
treatment. Clin Cancer Res 2009;15:2920�
2926.
11 Hofhuis JG, Spronk PE, van Stel HF, Schrijvers
GJ, Rommes JH, Bakker J: The impact of
critical illness on perceived health-related
quality of life during ICU treatment, hospital
stay, and after hospital discharge: a longterm
follow-up study. Chest 2008;133:377�
385.
12 Guest JF, Panca M, Baeyens JP, de Man F,
Ljungqvist O, Pichard C, Wait S, Wilson L:
Health economic impact of managing patients
following a community-based diagnosis
of malnutrition in the UK. Clin Nutr 2011;
30:422�429.
13 Kyle UG, Morabia A, Slosman DO, Mensi N,
Unger P, Pichard C: Contribution of body
composition to nutritional assessment at
hospital admission in 995 patients: a controlled
population study. Br J Nutr 2001;86:
725�731.
14 Kondrup J, Allison SP, Elia M; Vellas B,
Plauth M: Educational and Clinical Practice
Committee, European Society of Parenteral
and Enteral Nutrition (ESPEN): ESPEN
guidelines for nutrition screening 2002. Clin
Nutr 2003;22:415�421.
15 Haute Autorit� de Sant�: IPAQSS: informations.
2010. www.has-sante.fr/portail/
jcms/c_970427/ipaqss-informations.
16 World Health Organization: Obesity and
overweight: fact sheet No. 311. 2011. http://
www.who.int/mediacentre/factsheets/fs311/
en/index.html.
17 Thibault R, Chikhi M, Clerc A, Darmon P,
Chopard P, Picard-Kossovsky M, Genton L,
Pichard C: Assessment of food intake in hospitalised
patients: a 10 year-comparative
study of a prospective hospital survey. Clin
Nutr 2011;30:289�296.
18 Stenholm S, Harris TB, Rantanen T, Visser
M, Kritchevsky SB, Ferrucci L: Sarcopenic
obesity: definition, cause and consequences.
Curr Opin Clin Nutr Metab Care 2008;11:
693�700.
19 Pichard C, Kyle UG: Body composition measurements
during wasting diseases. Curr
Opin Clin Nutr Metab Care 1998;1:357�361.
20 Wang ZM, Pierson RN Jr, Heymsfield SB:
The five-level model: a new approach to organizing
body-composition research. Am J
Clin Nutr 1992;56:19�28.
21 Schols AM, Broekhuizen R, Weling-Scheepers
CA, Wouters EF: Body composition and
mortality in chronic obstructive pulmonary
disease. Am J Clin Nutr 2005;82:53�59.
22 Slinde F, Gronberg A, Engstrom CP, Rossander-Hulthen
L, Larsson S: Body composition
by bioelectrical impedance predicts
mortality in chronic obstructive pulmonary
disease patients. Respir Med 2005;99:1004�
1009.
23 Vestbo J, Prescott E, Almdal T, Dahl M, Nordestgaard
BG, Andersen T, Sorensen TI,
Lange P: Body mass, fat-free body mass, and
prognosis in patients with chronic obstructive
pulmonary disease from a random population
sample: findings from the Copenhagen
City Heart Study. Am J Respir Crit Care
Med 2006;173:79�83.
24 Segall L, Mardare NG, Ungureanu S, Busuioc
M, Nistor I, Enache R, Marian S, Covic A:
Nutritional status evaluation and survival in
haemodialysis patients in one centre from
Romania. Nephrol Dial Transplant 2009;24:
2536�2540.
25 Beddhu S, Pappas LM, Ramkumar N, Samore
M: Effects of body size and body composition
on survival in hemodialysis patients. J
Am Soc Nephrol 2003;14:2366�2372.
26 F�rstenberg A, Davenport A: Assessment
of body composition in peritoneal dialysis
patients using bioelectrical impedance and
dual-energy X-ray absorptiometry. Am J
Nephrol 2011;33:150�156.
27 Futter JE, Cleland JG, Clark AL: Body mass
indices and outcome in patients with chronic
heart failure. Eur J Heart Fail 2011;13:207�
213.
28 Marin B, Desport JC, Kajeu P, Jesus P, Nicolaud
B, Nicol M, Preux PM, Couratier P: Alteration
of nutritional status at diagnosis is
a prognostic factor for survival of amyotrophic
lateral sclerosis patients. J Neurol
Neurosurg Psychiatry 2011;82:628�634.
29 Janiszewski PM, Oeffinger KC, Church TS,
Dunn AL, Eshelman DA, Victor RG, Brooks
S, Turoff AJ, Sinclair E, Murray JC, Bashore
L, Ross R: Abdominal obesity, liver fat, and
muscle composition in survivors of childhood
acute lymphoblastic leukemia. J Clin
Endocrinol Metab 2007;92:3816�3821.
30 Wagner D, Adunka C, Kniepeiss D, Jakoby
E, Schaffellner S, Kandlbauer M, Fahrleitner-Pammer
A, Roller RE, Kornprat P, M�ller
H, Iberer F, Tscheliessnigg KH: Serum albumin,
subjective global assessment, body
mass index and the bioimpedance analysis in
the assessment of malnutrition in patients up
to 15 years after liver transplantation. Clin
Transplant 2011;25:E396�E400.
31 Kimyagarov S, Klid R, Levenkrohn S, Fleissig
Y, Kopel B, Arad M, Adunsky A: Body
mass index (BMI), body composition and
mortality of nursing home elderly residents.
Arch Gerontol Geriatr 2010;51:227�230.
32 Buffa R, Mereu RM, Putzu PF, Floris G,
Marini E: Bioelectrical impedance vector
analysis detects low body cell mass and dehydration
in patients with Alzheimer�s disease.
J Nutr Health Aging 2010;14:823�827.
33 Schols AM, Wouters EF, Soeters PB, Westerterp
KR: Body composition by bioelectricalimpedance
analysis compared with deuterium
dilution and skinfold anthropometry in
patients with chronic obstructive pulmonary
disease. Am J Clin Nutr 1991;53:421�424.
34 Thibault R, Le Gallic E, Picard-Kossovsky
M, Darmaun D, Chambellan A: Assessment
of nutritional status and body composition
in patients with COPD: comparison of several
methods (in French). Rev Mal Respir
2010;27:693�702.
35 Kyle UG, Janssens JP, Rochat T, Raguso CA,
Pichard C: Body composition in patients
with chronic hypercapnic respiratory failure.
Respir Med 2006;100:244�252.
36 Rieken R, van Goudoever JB, Schierbeek H,
Willemsen SP, Calis EA, Tibboel D, Evenhuis
HM, Penning C: Measuring body composition
and energy expenditure in children with
severe neurologic impairment and intellectual
disability. Am J Clin Nutr 2011;94:759�
766
37 Avram MM, Fein PA, Borawski C, Chattopadhyay
J, Matza B: Extracellular mass/body
cell mass ratio is an independent predictor of
survival in peritoneal dialysis patients. Kidney
Int Suppl 2010;117:S37�S40.
38 Frisancho AR: New norms of upper limb fat
and muscle areas for assessment of nutritional
status. Am J Clin Nutr 1981;34:2540�2545.
39 Caregaro L, Alberino F, Amodio P, Merkel C,
Bolognesi M, Angeli P, Gatta A: Malnutrition
in alcoholic and virus-related cirrhosis.
Am J Clin Nutr l996;63:602�609.
40 Alberino F, Gatta A, Amodio P, Merkel C, Di
Pascoli L, Boffo G, Caregaro L: Nutrition and
survival in patients with liver cirrhosis. Nutrition
2001;17:445�450.
41 Liu E, Spiegelman D, Semu H, Hawkins C,
Chalamilla G, Aveika A, Nyamsangia S,
Mehta S, Mtasiwa D, Fawzi W: Nutritional
status and mortality among HIV-infected
patients receiving antiretroviral therapy in
Tanzania. J Infect Dis 2011;204:282�290.
42 Soler-Cataluna JJ, Sanchez-Sanchez L, Martinez-Garcia
MA, Sanchez PR, Salcedo E,
Navarro M: Mid-arm muscle area is a better
predictor of mortality than body mass index
in COPD. Chest 2005;128:2108�2115.
43 Marquis K, Debigar� R, Lacasse Y, LeBlanc P,
Jobin J, Carrier G, Maltais F: Midthigh muscle
cross-sectional area is a better predictor
of mortality than body mass index in patients
with chronic obstructive pulmonary
disease. Am J Respir Crit Care Med 2002;15;
166:809�813.
44 Kyle UG, Pirlich M, Lochs H, Schuetz T, Pichard
C: Increased length of hospital stay in
underweight and overweight patients at hospital
admission: a controlled population
study. Clin Nutr 2005;24:133�142.
45 Kyle UG, Bosaeus I, De Lorenzo AD, Deurenberg
P, Elia M, G�mez JM, Heitmann
BL, Kent-Smith L, Melchior JC, Pirlich M,
Scharfetter H, Schols AM, Pichard C, Composition
of the ESPEN Working Group. Bioelectrical
impedance analysis. 1. Review of
principles and methods. Clin Nutr 2004;23:
1226�1243.
46 Santarpia L, Marra M, Montagnese C, Alfonsi
L, Pasanisi F, Contaldo F: Prognostic
significance of bioelectrical impedance
phase angle in advanced cancer: preliminary
observations. Nutrition 2009;25:930�931.
47 Gupta D, Lammersfeld CA, Vashi PG, King
J, Dahlk SL, Grutsch JF, Lis CG: Bioelectrical
impedance phase angle in clinical practice:
implications for prognosis in stage IIIB and
IV non-small cell lung cancer. BMC Cancer
2009;9:37.
48 Gupta D, Lis CG, Dahlk SL, Vashi PG,
Grutsch JF, Lammersfeld CA: Bioelectrical
impedance phase angle as a prognostic indicator
in advanced pancreatic cancer. Br J
Nutr 2004;92:957�962.
49 Gupta D, Lammersfeld CA, Burrows JL,
Dahlk SL, Vashi PG, Grutsch JF, Hoffman S,
Lis CG: Bioelectrical impedance phase angle
in clinical practice: implications for prognosis
in advanced colorectal cancer. Am J Clin
Nutr 2004;80:1634�1638.
50 Paiva SI, Borges LR, Halpern-Silveira D, Assun��o
MC, Barros AJ, Gonzalez MC: Standardized
phase angle from bioelectrical impedance
analysis as prognostic factor for
survival in patients with cancer. Support
Care Cancer 2010;19:187�192.
51 Schwenk A, Beisenherz A, R�mer K, Kremer
G, Salzberger B, Elia M: Phase angle from
bioelectrical impedance analysis remains an
independent predictive marker in HIV-infected
patients in the era of highly active antiretroviral
treatment. Am J Clin Nutr 2000;
72:496�501.
52 Desport JC, Marin B, Funalot B, Preux PM,
Couratier P: Phase angle is a prognostic factor
for survival in amyotrophic lateral sclerosis.
Amyotroph Lateral Scler 2008;9:273�
278.
53 Wirth R, Volkert D, R�sler A, Sieber CC,
Bauer JM: Bioelectric impedance phase angle
is associated with hospital mortality of
geriatric patients. Arch Gerontol Geriatr
2010;51:290�294.
54 Mushnick R, Fein PA, Mittman N, Goel N,
Chattopadhyay J, Avram MM: Relationship
of bioelectrical impedance parameters to nutrition
and survival in peritoneal dialysis patients.
Kidney Int Suppl 2003;87:S53�S56.
55 Selberg O, Selberg D: Norms and correlates
of bioimpedance phase angle in healthy human
subjects, hospitalized patients, and patients
with liver cirrhosis. Eur J Appl Physiol
2002;86:509�516.
56 Shah S, Whalen C, Kotler DP, Mayanja H,
Namale A, Melikian G, Mugerwa R, Semba
RD: Severity of human immunodeficiency
virus infection is associated with decreased
phase angle, fat mass and body cell mass in
adults with pulmonary tuberculosis infection
in Uganda. J Nutr 2001;131:2843�2847.
57 Barbosa-Silva MC, Barros AJ: Bioelectric impedance
and individual characteristics as
prognostic factors for post-operative complications.
Clin Nutr 2005;24:830�838.
58 Durnin JV, Womersley J: Body fat assessed
from total body density and its estimation
from skinfold thickness: measurements on
481 men and women aged from 16 to 72
years. Br J Nutr 1974;32:77�97.
59 Hill GL: Body composition research: implications
for the practice of clinical nutrition.
JPEN J Parenter Enter Nutr 1992;16:197�218.
60 Pierson RN Jr, Wang J, Thornton JC, Van
Itallie TB, Colt EW: Body potassium by fourpi
40K counting: an anthropometric correction.
Am J Physiol 1984;246:F234�F239.
61 Sohlstr�m A, Forsum E: Changes in total
body fat during the human reproductive cycle
as assessed by magnetic resonance imaging,
body water dilution, and skinfold thickness:
a comparison of methods. Am J Clin
Nutr 1997;66:1315�1322.
62 Leonard CM, Roza MA, Barr RD, Webber
CE: Reproducibility of DXA measurements
of bone mineral density and body composition
in children. Pediatr Radiol 2009;39:148�
154.
63 Genton L, Karsegard VL, Zawadynski S, Kyle
UG, Pichard C, Golay A, Hans DB: Comparison
of body weight and composition measured
by two different dual energy X-ray absorptiometry
devices and three acquisition
modes in obese women. Clin Nutr 2006;25:
428�437.
64 Jaffrin MY: Body composition determination
by bioimpedance: an update. Curr Opin
Clin Nutr Metab Care 2009;12:482�486.
65 Kyle UG, Pichard C, Rochat T, Slosman DO,
Fitting JW, Thiebaud D: New bioelectrical
impedance formula for patients with respiratory
insufficiency: comparison to dual-energy
X-ray absorptiometry. Eur Respir J 1998;
12:960�966.
66 Kyle UG, Bosaeus I, De Lorenzo AD, Deurenberg
P, Elia M, Manuel G�mez J, Lilienthal
Heitmann B, Kent-Smith L, Melchior
JC, Pirlich M, Scharfetter H, Schols AMWJ,
Pichard C, ESPEN: Bioelectrical impedance
analysis. 2. Utilization in clinical practice.
Clin Nutr 2004;23:1430�1453.
67 Mourtzakis M, Prado CM, Lieffers JR, Reiman
T, McCargar LJ, Baracos VE: A practical
and precise approach to quantification of
body composition in cancer patients using
computed tomography images acquired during
routine care. Appl Physiol Nutr Metab
2008;33:997�1006.
68 Bolton CE, Ionescu AA, Shiels KM, Pettit RJ,
Edwards PH, Stone MD, Nixon LS, Evans
WD, Griffiths TL, Shale DJ: Associated loss
of fat-free mass and bone mineral density in
chronic obstructive pulmonary disease. Am
J Respir Crit Care Med 2004;170:1286�1293.
69 Kyle UG, Genton L, Karsegard L, Slosman
DO, Pichard C: Single prediction equation
for bioelectrical impedance analysis in
adults aged 20�94 years. Nutrition 2001;17:
248�253.
70 Kyle UG, Genton L, Slosman DO, Pichard C:
Fat-free and fat mass percentiles in 5,225
healthy subjects aged 15 to 98 years. Nutrition
2001;17(7�8):534�541.
71 Kotler DP, Burastero S, Wang J, Pierson RN
Jr: Prediction of body cell mass, fat-free
mass, and total body water with bioelectrical
impedance analysis: effects of race, sex, and
disease. Am J Clin Nutr 1996;64:489S�497S.
72 Kyle UG, Genton L, Mentha G, Nicod L, Slosman
DO, Pichard C: Reliable bioelectrical
impedance analysis estimate of fat-free mass
in liver, lung, and heart transplant patients.
JPEN J Parenter Enteral Nutr 2001;25:45�51.
73 Mattar L, Godart N, Melchior JC, Falissard
B, Kolta S, Ringuenet D, Vindreau C, Nordon
C, Blanchet C, Pichard C: Underweight
patients with anorexia nervosa: comparison
of bioelectrical impedance analysis using
five equations to dual X-ray absorptiometry.
Clin Nutr 2011, E-pub ahead of print.
74 Genton L, Karsegard VL, Kyle UG, Hans DB,
Michel JP, Pichard C: Comparison of four
bioelectrical impedance analysis formulas in
healthy elderly subjects. Gerontology 2001;
47:315�323.
75 Robert S, Zarowitz BJ, Hyzy R, Eichenhorn
M, Peterson EL, Popovich J Jr: Bioelectrical
impedance assessment of nutritional status
in critically ill patients. Am J Clin Nutr 1993;
57:840�844.
76 Pichard C, Kyle UG, Slosman DO, Penalosa
B: Energy expenditure in anorexia nervosa:
can fat-free mass as measured by bioelectrical
impedance predict energy expenditure in
hospitalized patients? Clin Nutr 1996;15:
109�114.
77 Kreymann KG, Berger MM, Deutz NE, Hiesmayr
M, Jolliet P, Kazandjiev G, Nitenberg
G, van den Berghe G, Wernerman J, DGEM
(German Society for Nutritional Medicine),
Ebner C, Hartl W, Heymann C, Spies C, ESPEN:
ESPEN guidelines on enteral nutrition:
intensive care. Clin Nutr 2006;25:210�223.
78 Singer P, Berger MM, van den Berghe G, Biolo
G, Calder P, Forbes A, Griffiths R, Kreyman
G, Leverve X, Pichard C, ESPEN: ESPEN
guidelines on parenteral nutrition: intensive
care. Clin Nutr 2009;28:387�400.
79 Magnuson B, Peppard A, Auer Flomenhoft
D: Hypocaloric considerations in patients
with potentially hypometabolic disease
states. Nutr Clin Pract 2011;26:253�260.
80 Rigaud D, Boulier A, Tallonneau I, Brindisi
MC, Rozen R: Body fluid retention and body
weight change in anorexia nervosa patients
during refeeding. Clin Nutr 2010;29:749�
755.
81 Kyle UG, Chalandon Y, Miralbell R, Karsegard
VL, Hans D, Trombetti A, Rizzoli R,
Helg C, Pichard C: Longitudinal follow-up of
body composition in hematopoietic stem cell
transplant patients. Bone Marrow Transplant
2005;35:1171�1177.
82 Pison CM, Cano NJ, Cherion C, Caron F,
Court-Fortune I, Antonini M, GonzalezBermejo
J, Meziane L, Molano LC, Janssens
JP, Costes F, Wuyam B, Similowski T, Melloni
B, Hayot M, Augustin J, Tardif C,
Lejeune H, Roth H, Pichard C, the IRAD Investigators:
Multimodal nutritional rehabilitation
improves clinical outcomes of malnourished
patients with chronic respiratory
failure: a controlled randomised trial. Thorax
2011;66:953�960.
83 Pichard C, Kyle U, Chevrolet JC, Jolliet P,
Slosman D, Mensi N, Temler E, Ricou B: Lack
of effects of recombinant growth hormone
on muscle function in patients requiring
prolonged mechanical ventilation: a prospective,
randomized, controlled study. Crit
Care Med 1996;24:403�413.
84 Pichard C, Kyle UG, Jolliet P, Slosman DO,
Rochat T, Nicod L, Romand J, Mensi N,
Chevrolet JC: Treatment of cachexia with recombinant
growth hormone in a patient before
lung transplantation: a case report. Crit
Care Med 1999;27:1639�1642.
85 Leslie WD, Miller N, Rogala L, Bernstein
CN: Body mass and composition affect bone
density in recently diagnosed inflammatory
bowel disease: the Manitoba IBD Cohort
Study. Inflamm Bowel Dis 2009;15:39�46.
86 van der Meij BS, Langius JA, Smit EF,
Spreeuwenberg MD, von Blomberg BM,
Heijboer AC, Paul MA, van Leeuwen PA:
Oral nutritional supplements containing (n-
3) polyunsaturated fatty acids affect the nutritional
status of patients with stage III nonsmall
cell lung cancer during multimodality
treatment. J Nutr 2010;140:1774�1780.
87 Ryan AM, Reynolds JV, Healy L, Byrne M,
Moore J, Brannelly N, McHugh A, McCormack
D, Flood P: Enteral nutrition enriched
with eicosapentaenoic acid (EPA) preserves
lean body mass following esophageal cancer
surgery: results of a double-blinded randomized
controlled trial. Ann Surg 2009;249:
355�363.
88 Ndekha MJ, Oosterhout JJ, Zijlstra EE, Manary
M, Saloojee H, Manary MJ: Supplementary
feeding with either ready-to-use fortified
spread or corn-soy blend in wasted
adults starting antiretroviral therapy in Malawi:
randomised, investigator blinded, controlled
trial. BMJ 2009;338:b1867�b1875.
89 Ha L, Hauge T, Iversen PO: Body composition
in older acute stroke patients after treatment
with individualized, nutritional supplementation
while in hospital. BMC Geriatrics
2010;10:75.
90 Genton L, Karsegard VL, Chevalley T, Kossovsky
MP, Darmon P, Pichard C: Body
composition changes over 9 years in
healthy elderly subjects and impact of physical
activity. Clin Nutr 2011;30:436�442.
91 Reid CL, Murgatroyd PR, Wright A, Menon
DK: Quantification of lean and fat tissue repletion
following critical illness: a case report.
Crit Care 2008;12:R79.
92 Antoun S, Birdsel Ll, Sawyer MB, Venner P,
Escudier B, Baracos VE: Association of skeletal
muscle wasting with treatment with
sorafenib in patients with advanced renal
cell carcinoma: results from a placebo-controlled
study. J Clin Oncol 2010;28:1054�
1060.
93 Prado CM, Antoun S, Sawyer MB, Baracos
VE: Two faces of drug therapy in cancer:
drug-related lean tissue loss and its adverse
consequences to survival and toxicity. Curr
Opin Clin Nutr Metab Care 2011;14:250�
254.
94 Schutz Y, Kyle UG, Pichard C: Fat-free mass
index and fat mass index percentiles in Caucasians
aged 18�98 y. Int J Obes 2002;26:
953�960.
95 Kyle UG, Schutz Y, Dupertuis YM, Pichard
C: Body composition interpretation: contributions
of the fat-free mass index and the
body fat mass index. Nutrition 2003;19:597�
604.
96 Kyle UG, Piccoli A, Pichard C: Body composition
measurements: interpretation finally
made easy for clinical use. Curr Opin Clin
Nutr Metab Care 2003;6:387�393.

Close Accordion