twitter

Tuesday 4 December 2018

Integrative Medicine Chapter 88 - Antiinflammatory Diet

Integrative Medicine (Fourth Edition) 2018, Pages 869-877.e4 Author links open overlay panelWendyKohatsuMDScottKarpowiczMD https://doi.org/10.1016/B978-0-323-35868-2.00088-8 Get rights and content Previous chapter in book Next chapter in book Introduction The rate of chronic disease related to poor diet has risen to staggering proportions in the United States. A recent large meta-analysis indicates dietary factors are currently the leading risk factor for death and disability in the United States, responsible for more than 700,000 deaths and almost 15% of disability-adjusted life years in 2010.1 These diseases—including cardiovascular disease, diabetes, asthma, metabolic syndrome, depression, arthritis, and others—are characterized by a state of chronic, low-grade inflammation.2 Medications—nonsteroidal antiinflammatory drugs (NSAIDs), statins, and biological agents—certainly can be used to reduce inflammation;3 however, we must also help patients tackle the root causes of chronic inflammation created and perpetuated by a lifetime of unhealthy lifestyle habits. A large and growing body of evidence indicates that dietary modifications can have a significant impact on this chronic, low-grade inflammatory state.4, 5 Americans consume 5.5 pounds of food per day.6 We can take a more proactive, preventative approach by investing our 5+ pounds of daily food in healthy choices that decrease inflammation and thus chronic disease. To promote health and mitigate inflammation, integrative physicians must promote lifestyle changes and learn to use food as medicine. This chapter provides the most recent data to help integrative physicians tailor an antiinflammatory diet that is effective, acceptable, and delicious to each patient. Mechanisms and Measurement of Inflammation Several mechanisms by which food influences inflammation have been studied: • Modulation of eicosanoid activity. For example, compounds in culinary spices such as turmeric suppress cyclooxygenase-2 (COX-2) expression and in nutmeg, inhibit tumor necrosis factor (TNF)-alpha release in animal studies.7 • Pro- and antioxidant effects. Increased antioxidant food intake is correlated with lower C-reactive protein (CRP) levels8 and with a lower incidence of joint inflammation.9 • Insulin and glucose levels. A high dietary glycemic load was associated with elevated CRP concentrations,10 and elevation in inflammatory markers has been shown to precede type 2 diabetes11 (see Chapter 87). • Genetic and intracellular expression. A Mediterranean diet12 and omega-3 fats have been shown to modulate gene expression related to inflammation.13 • Modulation of endothelial function. Mediterranean diets powerfully affect endothelial markers.14 • Gastrointestinal dysbiosis. Numerous recent studies support that disruption to the gut microbiome can trigger a host of diverse inflammatory diseases.15, 16, 17 The gut microbiome may indeed be the frontier on which the battle of dietary inflammation is initially fought. Measuring Inflammation Current medical practice is better at diagnosing the multitude of illnesses caused by inflammation than catching it early in the disease process. Having validated ways to measure inflammation can enable us to intervene more proactively. CRP is a commonly used biomarker of inflammation, and CRP levels are elevated with both chronic disease and as an acute-phase reactant. High-sensitivity CRP (hs-CRP) is a more sensitive measure to assess basal levels of CRP at the lower range of 0.20–10.0 mg/L; an hs-CRP > 2.0 may signal higher cardiovascular risk.18 Other markers, such as TNF-alpha, interleukin-6 (IL-6), IL-1 beta, adhesion molecules, and endothelial function tests are used more widely in research to measure inflammation and are starting to emerge for clinical use. Recent studies have examined our food and its relationship with these inflammatory markers directly. A new dietary inflammatory index (DII)2 has been developed to assess the inflammatory potential of various diets, and a Mediterranean diet score is already in use.19 We anticipate that these tools will be useful for counseling patients on therapeutic diets in the future. The Mediterranean Diet Is an Archetype of an Antiinflammatory Diet The antiinflammatory diet is not one specific diet—indeed, there are many different diets that possess antiinflammatory effects. That being said, the Mediterranean diet is perhaps the most well-researched example of an antiinflammatory diet and serves as an archetypal model to further explore features of an antiinflammatory diet. Multiple studies and a recent meta-analysis have demonstrated the antiinflammatory effects of the Mediterranean diet as measured by serum inflammatory markers and measures of endothelial function.20, 21, 22 The importance of this diet is further underscored by the large and growing body of evidence from both epidemiological studies and randomized controlled trials that demonstrate a significant mortality benefit and reduction in chronic disease with adherence to the Mediterranean diet.19, 24, 25, 26 The Mediterranean diet is rich in vegetables, fruits, and whole grains; emphasizes nuts and olive oil as sources of fat; and gives preference to legumes, lean poultry, and fish rather than red meat (Fig. 88.1 and Table 88.1). Sodas, sweets, and refined baked goods; processed meats; and butter or similar spreadable fats are discouraged. Wine in moderation is an important component of the diet for those who choose to drink alcohol. This diet thus stands in stark contrast to the standard American diet, with its heavy intake of refined carbohydrates, red meat, added sugars, and limited portions of green vegetables, fruits, and whole grains. Download full-size image FIG. 88.1. The Mediterranean diet pyramid. TABLE 88.1. Original Mediterranean Diet Score19 Give yourself 1 point for each “yes” answer and 0 for each “no” Dietary Component YES NO Vegetables: 4 or more servings a day Legumes: 1 or more servings a week Fruit: 3 or more servings a day Nuts and seeds: 1 or more servings a week Whole grains: 1 or more servings a day Fish: 4 or more servings a week Fats: More unsaturated fats, such as olive oil, than saturated fats, such as butter Alcohol: 1/2 to 1 drink a day for women; 1 to 2 for men Red and processed meat: Fewer than 2 servings a day for women, fewer than 3 a day for men TOTAL: Totals of 6 or higher put you in the range of highest benefit. Scores less than 4 mean you are getting little or no protection. It is important to note that the Mediterranean diet is a pattern of eating, not a collection of specific recipes. Thus, while the traditional Mediterranean diet finds its origin in the Mediterranean region, the dietary pattern itself is broadly applicable across other regions, cultures, and food traditions. Multiple studies have demonstrated significant benefits of the Mediterranean diet when taken out of its traditional cultural context, including studies on Nordic,27 Polish,28 Indian,29 and Okinawan30 diets that follow the overall Mediterranean dietary pattern. Thus, patients whose individual food preferences may not match those of the Mediterranean region should have little difficulty adapting it successfully to their own taste preferences. It is also cost-effective.31, 32 While it is useful to explore in more detail the individual components of the Mediterranean diet, it is also important to note that the overall benefit of the Mediterranean diet is likely a result of the diet as a whole rather than any specific component. For example, in one of the most important epidemiological studies on the Mediterranean diet, the authors demonstrate that while an increase in a single component of the Mediterranean diet did not result in a significant benefit, an increase in any two components resulted in a statistically significant 25% reduction in overall mortality.19 Furthermore, data indicate that even relatively small changes can have important health impacts. The PREDIMED trial, a recent large randomized controlled trial assessing the impact of the Mediterranean diet on cardiovascular disease, showed that patients who were more adherent to the Mediterranean diet by just two of the component items significantly reduced their rate of cardiovascular disease and its associated mortality.23 Components of an Antiinflammatory Diet Fats Famous for their taste and palatability, antiinflammatory diets, especially the traditional Mediterranean diet, are characterized by their typically higher percentage of calories from fat—usually in the 40%–50% range—which adds both high satiety and flavor. Choosing the right fats—both the quantity as well as the quality—is important for reaping the key benefits of an antiinflammatory diet (Table 88.2). TABLE 88.2. The Skinny on Fats Type Subtype Metabolite Food Sources Notes Saturated Long chain TG Animal fats—meat, butter, cheese Amount in the diet is controversial, 7%–10% of calories. MCT SCT Tropical oils—coconut, palm Shorter-chain saturated fats may burn in the body more like carbohydrates. Monounsaturated Olive oil Nuts Avocados Peanuts Higher EVOO intake associated with a reduction in the CVD mortality by half.55 Reduces LDL and increases HDL. Polyunsaturated Omega-6 Essential fatty acid AA GLA Corn oil Most vegetable oils Soya Omega-6 fatty acids are essential fatty acids easily obtainable in the diet; however, usually in gross excess of omega-3s. Omega-3 Essential fatty acid ALA Flax, hemp, chia, walnuts ALA conversion to biologically active EPA and DHA is only about 10%–15%. EPA DHA Cold water fish (e.g., salmon), algae Potent antiinflammatory effects. Eat 2+ fatty fish servings/week.162 AA, arachidonic acid; ALA, alpha linolenic acid; DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; GLA, gamma linoleic acid, an antiinflammatory omega-6; MCT, medium-chain TG; SCT, short-chain TG; TG, triglyceride. a Soy contains both omega-6 (54%) and omega-3 (7%) fatty acids. There is a lot of controversy on the amounts of fats in a healthy diet. On one end of the spectrum, the Japanese diet contains only about 7% of its calories from fat, while a Mediterranean diet about 45%. For all types of fats, it is critical to pay attention to the quality, not just quantity. Choosing fresh, natural, organic, unrefined oils and fats will ensure protection against rancidity, oxidation, nutrient degradation, and contamination. The two essential fatty acids—omega-6 and omega-3—need to obtained from the diet as they cannot be synthesized by the body. In general, omega-6 fats lead to more inflammation, and omega-3 metabolism leads to the formation of less-inflammatory or antiinflammatory prostaglandins and leukotrienes. Thus, it is critical to keep a healthy balance between the two. A truly free-range egg has a favorably low omega-6:omega-3 ratio of only 1.3 compared to a conventionally raised egg with a ratio of 19.4.35 A few dietary fat dilemmas are as follows: 1. Omega-6 fats are vastly more abundant in the Western diet (usually 15–20×) compared with omega-3s. A ratio of 1:1–4:1 of omega-6 to omega-3 is recommended.36, 37 This means that Americans need to cut down on excessive intake of omega-6 fats and ramp up consumption of omega-3s (Fig. 88.2). Download full-size image FIG. 88.2. Influence of omega-6 and omega-3 fatty acids on inflammation. 2. Omega-3 and -6 fats compete for the same metabolic enzymes, so high levels of omega-6 fats may suppress the already limited metabolism of omega-3 fats. At low levels of omega-3 intake, omega-6 fats are associated with the highest amount of inflammation.38 3. Renewable, plant-based forms of omega-3 fats largely contain alpha-linolenic acid (ALA) rather than eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). The body needs to metabolize this ALA into EPA and DHA to exert its antiinflammatory effects, though it is able to do so in only relatively low amounts—only about 10%–15% of ALA is metabolized into EPA and then subsequently into DHA. Cold-water fatty fish, on the other hand, naturally contain higher amounts of preformed EPA and DHA, which exert antiinflammatory effects without the need for further metabolism. However, for environmental, heavy metal contamination, and cost issues, fish consumption is often limited. 4. Fish oil supplements do not seem to yield the same omega-3 benefits as eating fish itself.39, 40 Dose, quality, and human genetic variation may explain these differences.41 Remember the acronym S.M.A.S.H. for your best sources of omega-3 EPA and DHA in cold-water fish such as Salmon (wild Pacific), Mackerel (Spanish), Anchovies, Sardines, and Herring. The Evil of Trans-Fatty Acids When a naturally liquid polyunsaturated fat is chemically converted via hydrogenation to a more solid fat, synthetic trans-fats are formed (Fig. 88.3). While more shelf-stable, these trans-fats (also known as hydrogenated fats, or partially hydrogenated fats) wreak havoc on the body, disrupting the native cis-alignment of membrane fats,42 increasing serum levels of lipoprotein(a) and triglycerides,43 as well as inflammatory mediators. In June 2015, the U.S. Food and Drug Administration (FDA) finally declared that partially hydrogenated oils are not “generally regarded as safe” (GRAS) in human food. Manufacturers have 3 years to remove partially hydrogenated oils from their products. Download full-size image FIG. 88.3. Hydrogenation of polyunsaturated fat. Olive Oil Olive oil is a key component of the traditional Mediterranean diet and a growing body of evidence supports its inclusion as an important part of an antiinflammatory diet.14 Olive oil is rich in oleic acid, a monounsaturated fatty acid. Additionally, virgin and extra virgin olive oils, unrefined versions made by pressing freshly picked olives, have high quantities of polyphenols, which have potent antioxidative properties.45, 46 The benefits of olive oil likely derive from a synergistic relationship between these key components.47 Growing evidence from intervention-based trials indicates that olive oil reduces key cardiovascular risk factors. Olive oil, particularly the high-polyphenol virgin or extra virgin varieties, improves lipid profiles and reduces dyslipidemia through an improvement in the quantity and function of high-density lipoprotein (HDL) cholesterol and reduction in low-density lipoprotein (LDL) cholesterol.23, 48, 49 Additionally, olive oil has been shown to decrease blood pressure in patients with hypertension, improve endothelial function in patients with atherosclerotic disease, and reduce markers of inflammation.51, 52 Further evidence suggests that regular olive oil consumption, independent of total energy intake, decreases the risk of new-onset diabetes.53 Importantly, evidence from both epidemiological and intervention studies demonstrate a mortality benefit from olive oil consumption. The recent PREDIMED randomized controlled trial demonstrated a significant reduction in cardiovascular events and cardiovascular disease-related mortality in patients consuming a Mediterranean diet supplemented with extra virgin olive oil.23 Further analysis of these data suggests a 10% reduction in cardiovascular disease for every 10 additional grams of extra virgin olive oil consumed per day.55 These data correspond with multiple epidemiological studies that support a significant inverse correlation between olive oil, particularly the extra virgin variety, and coronary heart disease,56, 57 stroke,58 and cardiovascular mortality.59 Nuts Nuts are a rich source of monounsaturated and polyunsaturated fats, in addition to providing fiber, minerals, and other important nutrients.60, 61 Increased nut consumption has an important effect on reducing serum inflammatory markers, including CRP and IL-6.62 More importantly, multiple recent prospective studies indicate that increased nut consumption decreases metabolic syndrome,63 coronary artery disease,64 and total mortality in addition to cancer-specific and cardiovascular disease-specific mortality.65, 66 These studies support a dose–response relationship, and data from the PREDIMED trial suggest that consuming just three servings of nuts per week can reduce all-cause mortality by 39%.66 Despite slightly different fatty acid compositions and nutritional components, similar benefits are observed across a wide range of tree nuts as well as peanuts.65 Coconut Oil Recently, coconut oil has garnered considerable attention in the lay press as a health food with a myriad of purported health benefits.69 Saturated fat comprises more than 90% of the fat content of coconut oil, the majority of which is in the form of medium-chain fatty acids.70 However, in contrast to the longer-chain saturated fatty acids more typically found in animal fats, medium-chain fatty acids are more easily metabolized.71 Recent data from a human trial suggest that coconut oil, at least in daily doses of 30 mL, may not have the same dyslipidemic properties and proinflammatory properties as other saturated fats.72 In addition, virgin coconut oil contains high concentrations of polyphenols and may therefore have antioxidant as well as antiinflammatory effects, as demonstrated in animal models.73, 74 Nonetheless, there is a dearth of quality evidence from human trials assessing the antiinflammatory or overall health benefits of coconut oil, and additional data are needed before it can be strongly recommended as a component of an antiinflammatory diet. Carbohydrates Whole grains contain naturally occurring fiber, lignans, magnesium, zinc, B vitamins, and vitamin E that may help fight inflammation. Actively replacing refined carbs with whole grains in the diet has been shown to reduce inflammatory markers.75 Prospective studies have shown that higher dietary fiber and whole-grain intake is associated with decreased hs-CRP and IL-6.76, 77, 78 Whole-grain consumption substantially lowers the risk of inflammation-mediated diseases,79 such as cardiovascular disease, diabetes, cancer, and obesity. One study80 of 522 diabetic patients showed that including 15 g of dietary fiber for every 1000 calories consumed daily with lifestyle changes (moderate exercise) significantly reduced CRP by 27%. Perhaps the most important factor that best represents the inflammatory potential of a carbohydrate-containing food is its glycemic load (GL) (see Chapter 87). Excessive consumption of highly processed carbohydrates, e.g., white flour and refined sugars, with high GL, cause abnormal surges in blood glucose and insulin levels, which then overloads mitochondrial capacity thus creating free radicals.81 Immediate increases in CRP and inflammatory cytokines can be seen with a single meal.82 One study found a strong link between high dietary GL and elevated CRP concentrations in women; CRP levels were more than doubled (3.7 mg/L) in the group consuming the highest GL compared to the lowest GL (1.6 mg/L).10 Similarly, higher dietary glycemic index (GI)/GL has been associated with higher levels of TNF-alpha and IL-6.84 Even with the same amount of calories, choosing higher-quality carbohydrates with low GLs can lower the inflammatory profile.85 Not surprisingly, diets rich in unprocessed, natural plant foods such as the Okinawan or Mediterranean diets have lower GLs, substantially lower postprandial glucose levels, and are associated with improved cardiovascular health and longevity.86 Minimally processed low-GL foods—vegetables, fruit, nuts, seeds, and whole grains—do not result in adverse postprandial inflammatory effects, but high-GL foods do.87 Other mechanisms besides GL also likely help explain the benefit of whole grains in an antiinflammatory diet. A distinct benefit of whole-grain intake is supporting a more favorable gut microbiome composition that lowers both gut and systemic inflammation.88, 89, 90 Furthermore, another reason to consume whole grains is that the active phytochemicals are concentrated in bran (fiber) and the germ (carbohydrate); refining wheat with removal of the fiber causes a 200- to 300-fold loss in phytochemicals and increases the speed of sugar absorption (the GI)91 (Fig. 88.4)! Portion control is also important! Overconsumption of even healthy, low-GL foods can trigger a hyperglycemic response and inflammation. Excess calories regardless of source also contribute to obesity, which itself produces inflammation.92 Download full-size image FIG. 88.4. Fiber (bran) helps reduce the speed of sugar (germ) absorption. Vegetables and Fruit Vegetables and fruit—while often classified under the macronutrient “carbohydrate”—deserve their own stand-alone category due to their natural abundance of nutraceutical benefits! Vegetables contain the highest concentrations of vitamins, minerals, and other protective phytochemicals, with a lower caloric density compared with other foods. Rich in biochemical complexity, whole vegetables and fruit are superior to any single isolated nutrient. Citrus fruit, for example, contains not just vitamin C, but some 60 flavonoids, 20 carotenoids, and limonoids. Higher intake of vitamin C from food sources has been associated with lower CRP and tissue plasminogen activator (tPA) levels.93 The inadequate consumption of vegetables and fruit in the United States is appalling. It is estimated that Americans only consume 1.6 servings of vegetables per day and only one fruit per day.94 Fewer than 10% of Americans eat enough fruits and vegetables. Even more concerning is that the largest single contributor to fruit intake was orange juice and that potatoes still dominate vegetable consumption; fried potatoes account for about one-third of the “vegetables” that adolescents consume.95 Studies support that people who consume more vegetables and fruit have lower rates of inflammatory disease such as heart disease, insulin resistance, and some cancers.96, 97, 98, 99 High intake of vegetables and fruits, greater than five servings per day, has a significant inverse dose–response association with inflammatory markers such as CRP, IL-6, and adhesion factors.97, 100, 101 The more vegetables and fruit one eats, the less inflammation. Estimates from the US Department of Agriculture (USDA) flavonoid databases also show an inverse association between high flavonoid intake (from fruit and vegetables) and lower CRP levels.103 Flavonoids are responsible for the deep colors of fruits and vegetables and often are a proxy marker for high nutritional quality. In epidemiological studies, higher fruit and vegetable intake was shown to significantly decrease markers of inflammation in adolescents104 and in adult women.97 Adding fruits and vegetables to the diet, especially those rich in flavonoids, significantly improved microvascular reactivity and lowered CRP values.106 Watch the sugar! The American Heart Association recommends that women consume no more than 100 calories/day as added sugar. This equals 25 g of sugar or about six teaspoons. For men, the limit is 150 calories, 36 g of sugar, or nine teaspoons per day. Read labels, this adds up quickly! To satisfy your sweet tooth, choose fresh fruit in season.107 Proteins A large body of data supports the health benefits of plant-based proteins, and they are a healthy alternative to animal sources of protein.108, 109 Legumes are high in protein as well as fiber, iron, folic acid, and B vitamins. Most legumes are deficient in the essential amino acids methionine and tryptophan, but these are found in sufficient amounts in most grains. Randomized clinical trials indicate that regular consumption of legumes is associated with a reduction in inflammatory markers and an improvement in both lipid profiles and endothelial function.110, 111 Legumes make up a substantial part of a healthy antiinflammatory diet, can be prepared in many tasty ethnic dishes, and provide the majority of protein for a hungry world. Substituting healthy protein sources like legumes for red meat is associated with a lower mortality risk.112 In contrast, diets high in red and processed meats are correlated with increases in inflammatory markers, markers of glucose dysregulation, and dyslipidemia.113, 114, 115 Furthermore, diets higher in red meat, especially in processed forms (e.g., cured meats and sausages, bacon, and processed lunch meats) are associated with higher rates of cardiovascular disease, certain cancers, and all-cause mortality.116, 117, 118 Although it is not entirely clear what component of red meat contributes to its inflammatory effect, several clinical trials suggest that consuming lean meat may not result in the same proinflammatory effect that is observed with the higher fat, more processed red meats typically found in a modern American diet.119, 120, 121 For those who do eat meat, studies support trimming visible fat122 and avoiding charring the food to lessen the inflammatory impact.123 In addition to lean meats, animal protein sources from fish124 and dairy125, 126 are suitable components of an antiinflammatory diet. Fish While a source of high-quality protein and healthy fat, fish often swim in waters contaminated with heavy metals such as mercury, cadmium, lead, and other pollutants such as polychlorinated biphenyls (PCBs). These are proinflammatory and impair metabolic function. Even taking this into consideration, one review127 reports that the health benefits of eating fish exceed the potential risks. Avoid fish highest in methylmercury—shark, swordfish, golden bass (tilefish), and king mackerel. A serving of albacore tuna contains 0.35 ppm methylmercury with 1.0 ppm being the EPA’s “allowable” upper limit/day, so limit it to no more than one serving per week.128 Some of the safest fish are tilapia, anchovies, and wild salmon. Other Antiinflammatory Foods, Spices, and Supplements • Turmeric: Turmeric is the queen of antiinflammatory spices! Multiple studies have assessed the potential benefits of curcumin, the main component of turmeric (Curcuma longa), on a variety of inflammatory conditions. Several recent randomized controlled trials have shown improvements in inflammatory markers and endothelial function with regular administration of curcumin.129, 130, 131 Furthermore, trials have found curcumin equivalent in pain relief to ibuprofen and diclofenac for knee osteoarthritis132 and rheumatoid arthritis,133 respectively. Curcumin has shown promise in other inflammatory conditions, including maintenance of remission in ulcerative colitis134 and improvement in signs and symptoms of oral lichen planus.135 Especially considering its long track record of culinary use and excellent side effect profile in all the trials in which it has been studied, turmeric represents both a safe and effective addition to any antiinflammatory diet. • Wine: While excessive alcohol consumption is clearly linked to excess morbidity and mortality, a large body of epidemiological data suggests that low-to-moderate alcohol consumption is associated with decreased levels of both cardiovascular and all-cause mortality.136, 137, 138, 139 Wine is of particular interest in the antiinflammatory diet as it is rich in polyphenols, which likely exert numerous antioxidative and antiinflammatory effects.140, 141 Randomized trials have demonstrated a reduction in inflammatory markers142 and improvement in lipid profiles143 with regular, moderate consumption of red wine. • Other spices—ginger, chili pepper, oregano, basil, thyme, and rosemary—also possess antiinflammatory properties144, 145 and add flavor to a wide variety of cuisines. • Cocoa: Cocoa and cocoa-rich products such as dark chocolate contain high quantities of flavonoids, as well as fiber and other important nutrients.146 Multiple trials demonstrate a reduction in serum inflammatory markers,147, 148, 149 improvement in lipid profiles,150, 151, 152 and enhanced endothelial function153, 154, 155 with consumption of flavonoid-rich cocoa. Furthermore, a recent trial demonstrated improvement in both walking distance and time in patients with peripheral artery disease after consumption of 40 g daily of dark chocolate.156 Evidence from a meta-analysis of randomized controlled trials indicates a reduction in insulin resistance,157 and epidemiological data suggests a reduction in diabetes incidence with cocoa consumption.158 Mortality data are limited, though one epidemiological study indicated an inverse correlation between chocolate consumption and cardiovascular and all-cause mortality.159 Doses used in trials vary widely from less than 10 g daily to 100 g daily—although the lowest effective dose is not known, 1–2 ounces of dark (at least 70%) chocolate as a tasty and healthy treat is a safe suggestion. Medical Conditions That May Improve With an Antiinflammatory Diet Table 88.3 provides a summary of some of the major diseases for which antiinflammatory diet modifications have shown therapeutic promise. TABLE 88.3. Medical Conditions That May Improve With an Antiinflammatory Diet Arthritis Two randomized controlled trials found turmeric extracts to be as good or better than nonsteroidal antiinflammatory drugs (NSAIDs) at controlling symptoms of knee osteoarthritis132 and rheumatoid arthritis.133 Doses ranged from 500 mg to 1500 mg daily in these trials. Daily administration of a probiotic for 8 weeks was shown in a randomized controlled trial to reduce symptom severity and inflammatory status in patients with rheumatoid arthritis.165 Hypertension Randomized controlled trials found significant reductions in blood pressure in patients who adopted the traditional Mediterranean diet166 and the New Nordic Diet, a Mediterranean diet regional variant.27 Apart from its inclusion in the Mediterranean diet, daily consumption of olive oil results in a clinically significant reduction in blood pressure168—olive oil rich in polyphenols, such as extra virgin olive oil, may be more effective at decreasing blood pressure than regular olive oil.169 Olive oil dosing ranged from 30 mL–60 mL (2–4 tablespoons) daily in these trials. Asthma A recent small trial found improvement in spirometry and quality of life scores in patients with asthma who adopted a Mediterranean diet.170 COPD In one randomized controlled trial, patients with COPD showed significant improvement in lung function [increased forced expiratory volume in 1 second (FEV1)] with greater intake of antioxidant-rich fruits and vegetables.171 Diabetes Data from the PREDIMED trial indicate reduced onset of diabetes with increased adherence to an energy-unrestricted Mediterranean diet.53, 173 In patients with existing diabetes, adherence to an energy-unrestricted Mediterranean diet resulted in a reduction in hemoglobin A1c equivalent to that from mono-drug therapy.174 A low-carbohydrate Mediterranean diet has also been shown to reduce the need for antiglycemic medications in patients with newly diagnosed diabetes.175 Cardiovascular Disease Large randomized controlled trials have demonstrated the Mediterranean diet as an effective means of both primary23 and secondary26, 177 prevention of cardiovascular events, including myocardial infarction and stroke. Supplementation with olive oil55 and nuts66 likely provides even greater benefit. The effect size of dietary change should not be underestimated—secondary prevention trials indicated risk reductions of nearly two-thirds in those adhering to a Mediterranean-style diet. Peripheral Artery Disease The PREDIMED trial suggests that adherence to a Mediterranean diet significantly decreases the rate of new-onset symptomatic peripheral artery disease.181 In a randomized controlled trial of patients with existing peripheral artery disease, daily administration of 40 g of dark chocolate resulted in a significant improvement in walking autonomy.156 Obesity Adherence to a Mediterranean diet was associated with 51% lower odds of being obese and 59% lower odds of central obesity.183 A high amount of olive oil consumption was not associated with higher weight gain or a significantly higher risk of becoming overweight or obese.184 Despite a higher fat content (35% compared to 20%), participants eating a Mediterranean-based diet lost 4.1 kg compared with controls who gained 2.9 kg over 18 months.185 Inflammatory Bowel Disease A randomized controlled trial found that turmeric (Curcumin) at daily doses of 2 g given in conjunction with standard therapy significantly reduced rates of relapse and improved endoscopic and symptom-based disease severity scores.134 A recent case series demonstrated that patients who adhered to a modified antiinflammatory diet showed significant improvement in disease-related symptom scores.187 Nonalcoholic Fatty Liver Disease In adults with nonalcoholic fatty liver disease, data from a randomized controlled trial indicate that patients whose diets are supplemented with olive oil or canola oil at doses up to 20 mg daily showed a reduction in liver span and severity of fatty liver at 6 months.188 A recent meta-analysis found that daily supplementation with omega-3 fatty acids reduced hepatic steatosis.189 Daily doses above 800 mg are effective, although the ideal dose is not yet known. Cancer A recent meta-analysis found that higher adherence to the Mediterranean diet was associated with a 10% reduction in overall cancer incidence and mortality.190 This data indicate the effect may be most pronounced in colorectal and prostate cancer, with reductions in incidence of 14% and 4%, respectively, with increased adherence to the Mediterranean diet. Alzheimer’s Disease and Dementia Adherence to a Mediterranean diet supplemented with olive oil or nuts has been shown to improve cognition and slow age-related cognitive decline.191, 192 In an observational study, higher adherence to a Mediterranean diet was associated with slower cognitive decline as measured by the Mini Mental State Evaluation.193 A recent small pilot study found that daily supplementation with omega-3 fatty acids and alpha lipoic acid (ALA) reduced the progression of cognitive and functional decline in patients with Alzheimer’s disease.194 Doses used in this study were 1.6 g of omega-3 fatty acids and 600 mg of ALA daily. Depression A recent meta-analysis of 19 randomized controlled trials found omega-3 fatty acid supplementation effective in treating patients with both major depressive disorder (MDD) and depressive symptoms without a clear MDD diagnosis.195 Dosing varied widely across trials but averaged 1.4 g of combined EPA and DHA daily. Additionally, another recent trial found that initiating omega-3 supplementation with a selective serotonin reuptake inhibitor (SSRI) resulted in better reduction in depressive symptoms than the SSRI alone.196 Psoriasis In patients being treated for psoriasis, a randomized controlled trial demonstrated that a hypocaloric, omega-3 rich diet significantly reduced symptom severity at 3 and 6 months.197 Conclusion Inflammation is a common pathophysiological mechanism underlying most chronic disease. The antiinflammatory diet is based on evidence-based principles of sound eating to promote health and prevent and reduce inflammation in the body. It is whole food-based nutrition that emphasizes healthy fats, vegetables and fruit, whole grains, legumes, fish, and limited amounts of meat and dairy. It can be recommended as “food as medicine” for a wide variety of common diseases including heart disease, diabetes, obesity, cancer, Alzheimer’s disease, chronic obstructive pulmonary disease (COPD), depression, and gastrointestinal (GI) disorders. Key Web Resources Oldways Preservation Trust. Has the original Mediterranean diet pyramid, and also features culturally inclusive Asian, Latino, and African heritage pyramids. http://oldwayspt.org Anti-Inflammatory Food Pyramid. Dr. Andrew Weil has published a patient-friendly and illustrative antiinflammatory food pyramid. This plan also features berries, Asian mushrooms, soy, tea, and dark chocolate http://www.drweil.com and search for “food pyramid.”119 For data on mercury levels in fish, check out the U.S. Environmental Protection Agency. http://www.fda.gov/food/foodborneillnesscontaminants/metals/ucm115644.htm A good website with healthy Mediterranean-style recipes http://www.eatingwell.com/recipes_menus/collections/healthy_mediterranean_recipes Check out the “Meatless Monday” campaign online. This international movement encourages all to skip meat once a week and explore healthy and tasty vegetarian options. http://www.meatlessmonday.com Download full-size image FIG. 88.5. Patient handout: Antiinflammatory diet. References 1 C.J.L. Murray The state of US health, 1990-2010 JAMA, 310 (2013), p. 591 CrossRefView Record in ScopusGoogle Scholar 2 N. Shivappa, S.E. Steck, T.G. Hurley, et al. Designing and developing a literature-derived, population-based dietary inflammatory index Public Health Nutr, 17 (2014), pp. 1689-1696 CrossRefView Record in ScopusGoogle Scholar 3 P.M. Ridker, E. Danielson, F.A. Fonseca, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein N Engl J Med, 359 (2008), pp. 2196-2207 Google Scholar 4 P.P. Cavicchia, S.E. Steck, T.G. Hurley, et al. A new dietary inflammatory index predicts interval changes in serum high-sensitivity C-reactive protein J Nutr, 139 (2009), pp. 2365-2372 CrossRefView Record in ScopusGoogle Scholar 5 D. Giugliano, A. Ceriello, K. Esposito The effects of diet on inflammation. Emphasis on the metabolic syndrome J Am Coll Cardiol, 48 (2006), pp. 677-685 ArticleDownload PDFView Record in ScopusGoogle Scholar 6 DoSomething.org: 11 Facts about American eating habits: https://www.dosomething.org/facts/11-facts-about-american-eating-habits; Accessed July 15, 2015. Google Scholar 7 M.B. Augustine Integrative approach to nutrition B. Kligler, R. Lee (Eds.), Integrative medicine: principles for practice, McGraw Hill, New York (2004) Google Scholar 8 F. Brighenti, S. Valtuena, N. Pellegrini, et al. Total antioxidant capacity of the diet is inversely and independently related to plasma concentration of high-sensitivity C-reactive protein in adult Italian subjects Br J Nutr, 93 (2005), pp. 619-625 CrossRefView Record in ScopusGoogle Scholar 9 D.J. Pattison, D.P. Symmons, M. Lunt, et al. Dietary beta-cryptoxanthin and inflammatory polyarthritis: results from a population-based prospective study Am J Clin Nutr, 82 (2005), pp. 451-455 View Record in ScopusGoogle Scholar 10 S. Liu, J.E. Manson, J.E. Buring, et al. Relation between a diet with a high glycemic load and plasma concentrations of high-sensitivity C-reactive protein in middle-aged women Am J Clin Nutr, 75 (2002), pp. 492-498 View Record in ScopusGoogle Scholar 11 C. Herder, T. Illig, W. Rathmann, et al. Inflammation and type 2 diabetes: results from KORA Augsburg Gesundheitswesen, 67 (2005), pp. S115-S121 View Record in ScopusGoogle Scholar 12 V. Llorente-Cortés, R. Estruch, M.P. Mena, et al. Effect of Mediterranean diet on the expression of pro-atherogenic genes in a population at high cardiovascular risk Atherosclerosis, 208 (2010), pp. 442-450 ArticleDownload PDFView Record in ScopusGoogle Scholar 13 M. Massaro, E. Scoditti, M.A. Carluccio, et al. Omega-3 fatty acids, inflammation and angiogenesis: nutrigenomic effects as an explanation for anti-atherogenic and anti-inflammatory effects of fish and fish oils J Nutrigenet Nutrigenomics, 1 (2008), pp. 4-23 CrossRefView Record in ScopusGoogle Scholar 14 R. Casas, E. Sacanella, R. Estruch The immune protective effect of the Mediterranean diet against chronic low-grade inflammatory diseases Endocr Metab Immune Disord Drug Targets, 14 (2014), pp. 245-254 CrossRefView Record in ScopusGoogle Scholar 15 C.E. West, H. Renz, M.C. Jenmalm, et al. The gut microbiota and inflammatory no communicable diseases: associations and potentials for gut microbiota therapies J Allergy Clin Immunol, 135 (2015), pp. 3-13 ArticleDownload PDFView Record in ScopusGoogle Scholar 16 S.E. Jones, J. Versalovic Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors BMC Microbiol, 9 (2009), p. 35 CrossRefGoogle Scholar 17 J. Sun, E.B. Chang Exploring gut microbes in human health and disease: pushing the envelope Genes Dis, 1 (2014), pp. 132-139 ArticleDownload PDFView Record in ScopusGoogle Scholar 18 Mayo Medical Laboratories: C-reactive protein: http://www.mayomedicallaboratories.com/test-catalog/Clinical+and+Interpretive/82047. Accessed July 10, 2015. Google Scholar 19 A. Trichopoulou, T. Costacou, C. Bamia, et al. Adherence to a Mediterranean diet and survival in a Greek population N Engl J Med, 348 (2003), pp. 2599-2608 CrossRefView Record in ScopusGoogle Scholar 20 L. Schwingshackl, G. Hoffmann Mediterranean dietary pattern, inflammation and endothelial function: a systematic review and meta-analysis of intervention trials Nutr Metab Cardiovasc Dis, 24 (2014), pp. 929-939 ArticleDownload PDFView Record in ScopusGoogle Scholar 21 C. Richard, P. Couture, S. Desroches, et al. Effect of the Mediterranean diet with and without weight loss on markers of inflammation in men with metabolic syndrome Obesity, 21 (2013), pp. 51-57 CrossRefView Record in ScopusGoogle Scholar 22 K. Esposito, R. Marfella, M. Ciotola, et al. Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial JAMA, 292 (2004), pp. 1440-1446 CrossRefView Record in ScopusGoogle Scholar 23 R. Estruch, E. Ros, J. Salas-Salvadó, et al. Primary prevention of cardiovascular disease with a Mediterranean diet N Engl J Med, 368 (2013), pp. 1279-1290 CrossRefView Record in ScopusGoogle Scholar 24 Reference deleted in author review. Google Scholar 25 F. Sofi, R. Abbate, G.F. Gensini, et al. Accruing evidence on benefits of adherence to the Mediterranean diet on health: an updated systematic review and meta-analysis Am J Clin Nutr, 92 (2010), pp. 1189-1196 CrossRefView Record in ScopusGoogle Scholar 26 M. de Lorgeril, P. Salen, J.L. Martin, et al. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study Circulation, 99 (1999), pp. 779-785 CrossRefView Record in ScopusGoogle Scholar 27 S.K. Poulsen, A. Due, A.B. Jordy, et al. Health effect of the new Nordic diet in adults with increased waist circumference: a 6-mo randomized controlled trial Am J Clin Nutr, 99 (2014), pp. 35-45 CrossRefView Record in ScopusGoogle Scholar 28 G. Grosso, U. Stepaniak, A. Micek, et al. A Mediterranean-type diet is associated with better metabolic profile in urban Polish adults: results from the HAPIEE study Metabolism, 64 (2015), pp. 738-746 ArticleDownload PDFView Record in ScopusGoogle Scholar 29 R.B. Singh, G. Dubnov, M.A. Niaz, et al. Effect of an Indo-Mediterranean diet on progression of coronary artery disease in high risk patients (Indo-Mediterranean Diet Heart Study): a randomised single-blind trial Lancet, 360 (2002), pp. 1455-1461 ArticleDownload PDFView Record in ScopusGoogle Scholar 30 D.C. Willcox, B.J. Willcox, H. Todoriki, et al. The Okinawan diet: health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in glycemic load J Am Coll Nutr, 28 (Suppl) (2009), pp. 500S-516S CrossRefGoogle Scholar 31 K. Dalziel, L. Segal, M.A. de Lorgeril Mediterranean diet is cost-effective in patients with previous myocardial infarction J Nutr, 136 (2006), pp. 1879-1885 View Record in ScopusGoogle Scholar 32 K. Dalziel, L. Segal Time to give nutrition interventions a higher profile: cost-effectiveness of 10 nutrition interventions Health Promot Int, 22 (2007), pp. 271-283 CrossRefView Record in ScopusGoogle Scholar 33 Reference deleted in author review. Google Scholar 34 Reference deleted in author review. Google Scholar 35 A.P. Simopoulos Omega-3 fatty acids in health and disease and in growth and development Am J Clin Nutr, 54 (1991), pp. 438-463 CrossRefView Record in ScopusGoogle Scholar 36 A.P. Simopoulos Essential fatty acids in health and chronic disease Am J Clin Nutr, 70 (1999), pp. 560S-569S Google Scholar 37 A.P. Simopoulos Evolutionary aspects of diet: the omega-6/omega-3 ratio and the brain Mol Neurobiol, 44 (2011), pp. 203-215 CrossRefView Record in ScopusGoogle Scholar 38 T. Pischon, S.E. Hankinson, G.S. Hotamisligil, et al. Habitual dietary intake of n-3 and n-6 fatty acids in relation to inflammatory markers among US men and women Circulation, 108 (2003), pp. 155-160 CrossRefView Record in ScopusGoogle Scholar 39 R. Chowdhury, S. Warnakula, S. Kunutsor, et al. Association of dietary, circulating, and supplement fatty acids with coronary risk: a systematic review and meta-analysis Ann Intern Med, 160 (2014), pp. 398-406 CrossRefView Record in ScopusGoogle Scholar 40 A. Grey, M. Bolland Clinical trial evidence and use of fish oil supplements JAMA Intern Med, 174 (2014), pp. 460-462 CrossRefView Record in ScopusGoogle Scholar 41 H.R. Superko, A.R. Superko, G.P. Lundberg, et al. Omega-3 fatty acid blood levels clinical significance update Curr Cardiovasc Risk Rep, 8 (2014), p. 407 Google Scholar 42 U. Erasmus Fats that heal, fats that kill Alive Books, Summertown, TN (1993) Google Scholar 43 J.F. Mauger, A.H. Lichtenstein, L.J. Ausman, et al. Effect of different forms of dietary hydrogenated fats on LDL particle size Am J Clin Nutr, 78 (2003), pp. 370-375 View Record in ScopusGoogle Scholar 44 Reference deleted in author review. Google Scholar 45 E. Gimeno, A.I. Castellote, R.M. Lamuela-Raventós, et al. The effects of harvest and extraction methods on the antioxidant content (phenolics, α-tocopherol, and β-carotene) in virgin olive oil Food Chem, 78 (2002), pp. 207-211 ArticleDownload PDFView Record in ScopusGoogle Scholar 46 M.N. Franco, T. Galeano-Díaz, O. López, et al. Phenolic compounds and antioxidant capacity of virgin olive oil Food Chem, 163 (2014), pp. 289-298 ArticleDownload PDFView Record in ScopusGoogle Scholar 47 J. López-Miranda, F. Pérez-Jiménez, E. Ros, et al. Olive oil and health: summary of the II international conference on olive oil and health consensus report, Jaén and Córdoba (Spain) 2008 Nutr Metab Cardiovasc Dis, 20 (2010), pp. 284-294 ArticleDownload PDFView Record in ScopusGoogle Scholar 48 M.I. Covas, K. Nyyssönen, H.E. Poulsen, et al. The effect of polyphenols in olive oil on heart disease risk factors: a randomized trial Ann Intern Med, 145 (2006), pp. 333-341 CrossRefView Record in ScopusGoogle Scholar 49 Fernández-Castillejo S. Hernáez Á, M. Farràs, et al. Olive oil polyphenols enhance high-density lipoprotein function in humans: a randomized controlled trial Arterioscler Thromb Vasc Biol, 34 (2014), pp. 2115-2119 Google Scholar 50 Reference deleted in author review. Google Scholar 51 R. Moreno-Luna, R. Muñoz-Hernandez, M.L. Miranda, et al. Olive oil polyphenols decrease blood pressure and improve endothelial function in young women with mild hypertension Am J Hypertens, 25 (2012), pp. 1299-1304 View Record in ScopusGoogle Scholar 52 R.J. Widmer, M.A. Freund, A.J. Flammer, et al. Beneficial effects of polyphenol-rich olive oil in patients with early atherosclerosis Eur J Nutr, 52 (2013), pp. 1223-1231 CrossRefView Record in ScopusGoogle Scholar 53 J. Salas-Salvadó, M. Bulló, R. Estruch, et al. Prevention of diabetes with Mediterranean diets: a subgroup analysis of a randomized trial Ann Intern Med, 160 (2014), pp. 1-10 View Record in ScopusGoogle Scholar 54 Reference deleted in author review. Google Scholar 55 M. Guasch-Ferré, F.B. Hu, M.A. Martínez-González, et al. Olive oil intake and risk of cardiovascular disease and mortality in the PREDIMED study BMC Med, 12 (2014), p. 78 Google Scholar 56 G. Buckland, N. Travier, A. Barricarte, et al. Olive oil intake and CHD in the European prospective investigation into cancer and nutrition Spanish cohort Br J Nutr, 108 (2012), pp. 2075-2082 CrossRefView Record in ScopusGoogle Scholar 57 B. Bendinelli, G. Masala, C. Saieva, et al. Fruit, vegetables, and olive oil and risk of coronary heart disease in Italian women: the EPICOR Study Am J Clin Nutr, 93 (2011), pp. 275-283 CrossRefView Record in ScopusGoogle Scholar 58 C. Samieri, C. Féart, C. Proust-Lima, et al. Olive oil consumption, plasma oleic acid, and stroke incidence Neurology, 77 (2011), pp. 418-425 CrossRefView Record in ScopusGoogle Scholar 59 G. Buckland, A.L. Mayén, A. Agudo, et al. Olive oil intake and mortality within the Spanish population (EPIC-Spain) Am J Clin Nutr, 96 (2012), pp. 142-149 CrossRefView Record in ScopusGoogle Scholar 60 E. Ros, J. Mataix Fatty acid composition of nuts–implications for cardiovascular health Br J Nutr, 96 (Suppl 2) (2006), pp. S29-S35 CrossRefView Record in ScopusGoogle Scholar 61 R. Segura, C. Javierre, M.A. Lizarraga, et al. Other relevant components of nuts: phytosterols, folate and minerals Br J Nutr, 96 (Suppl 2) (2006), pp. S36-S44 CrossRefView Record in ScopusGoogle Scholar 62 R. Jiang, D.R. Jacobs Jr., E. Mayer-Davis, et al. Nut and seed consumption and inflammatory markers in the multi-ethnic study of atherosclerosis Am J Epidemiol, 163 (2006), pp. 222-231 CrossRefView Record in ScopusGoogle Scholar 63 S. Blanco Mejia, C.W. Kendall, E. Viguiliouk, et al. Effect of tree nuts on metabolic syndrome criteria: a systematic review and meta-analysis of randomised controlled trials BMJ Open, 4 (2014), p. e004660 CrossRefGoogle Scholar 64 L. Ma, F. Wang, W. Guo, et al. Nut consumption and the risk of coronary artery disease: a dose-response meta-analysis of 13 prospective studies Thromb Res, 134 (2014), pp. 790-794 ArticleDownload PDFView Record in ScopusGoogle Scholar 65 Y. Bao, J. Han, F.B. Hu, et al. Association of nut consumption with total and cause-specific mortality N Engl J Med, 369 (2013), pp. 2001-2011 CrossRefView Record in ScopusGoogle Scholar 66 M. Guasch-Ferré, M. Bulló, Martínez-González MÁ, et al. Frequency of nut consumption and mortality risk in the PREDIMED nutrition intervention trial BMC Med, 11 (2013), p. 164 Google Scholar 67 Reference deleted in author review. Google Scholar 68 Reference deleted in author review. Google Scholar 69 A.M. Marina, Y.B. Che Man, I. Amin Virgin coconut oil: emerging functional food oil Trends Food Sci Technol, 20 (2009), pp. 481-487 ArticleDownload PDFView Record in ScopusGoogle Scholar 70 J. Orsavova, L. Misurcova, J.V. Ambrozova, et al. Fatty acids composition of vegetable oils and its contribution to dietary energy intake and dependence of cardiovascular mortality on dietary intake of fatty acids Int J Mol Sci, 16 (2015), pp. 12871-12890 CrossRefView Record in ScopusGoogle Scholar 71 W.M. Fernando, I.J. Martins, K.G. Goozee, et al. The role of dietary coconut for the prevention and treatment of Alzheimer’s disease: potential mechanisms of action Br J Nutr, 114 (2015), pp. 1-14 CrossRefView Record in ScopusGoogle Scholar 72 M.L. Assunção, H.S. Ferreira, A.F. dos Santos, et al. Effects of dietary coconut oil on the biochemical and anthropometric profiles of women presenting abdominal obesity Lipids, 44 (2009), pp. 593-601 CrossRefView Record in ScopusGoogle Scholar 73 A. Vysakh, M. Ratheesh, T.P. Rajmohanan, et al. Polyphenolics isolated from virgin coconut oil inhibits adjuvant induced arthritis in rats through antioxidant and anti-inflammatory action Int Immunopharmacol, 20 (2014), pp. 124-130 ArticleDownload PDFView Record in ScopusGoogle Scholar 74 S.K. Yeap, B.K. Beh, N.M. Ali, et al. Antistress and antioxidant effects of virgin coconut oil in vivo Exp Ther Med, 9 (2015), pp. 39-42 CrossRefView Record in ScopusGoogle Scholar 75 P. Vitaglione, I. Mennella, R. Ferracane, et al. Whole-grain wheat consumption reduces inflammation in a randomized controlled trial on overweight and obese subjects with unhealthy dietary and lifestyle behaviors: role of polyphenols bound to cereal Am J Clin Nutr, 101 (2015), pp. 251-261 CrossRefView Record in ScopusGoogle Scholar 76 J. Goletzke, A.E. Buyken, G. Joslowski, et al. Increased intake of carbohydrates from sources with a higher glycemic index and lower consumption of whole grains during puberty are prospectively associated with higher IL-6 concentrations in younger adulthood among healthy individuals J Nutr, 144 (2014), pp. 1586-1593 CrossRefView Record in ScopusGoogle Scholar 77 O. Gögebakan, A. Kohl, M.A. Osterhoff, et al. Effects of weight loss and long-term weight maintenance with diets varying in protein and glycemic index on cardiovascular risk factors: the diet, obesity, and genes (DiOGenes) study: a randomized, controlled trial Circulation, 124 (2011), pp. 2829-2838 CrossRefView Record in ScopusGoogle Scholar 78 A.J. Gaskins, S.L. Mumford, A.J. Rovner, et al. Whole grains are associated with serum concentrations of high sensitivity C-reactive protein among premenopausal women J Nutr, 140 (2010), pp. 1669-1676 CrossRefView Record in ScopusGoogle Scholar 79 K. Pol, R. Christensen, E.M. Bartels, et al. Whole grain and body weight changes in apparently healthy adults: a systematic review and meta-analysis of randomized controlled studies Am J Clin Nutr, 98 (2013), pp. 872-884 CrossRefView Record in ScopusGoogle Scholar 80 C. Herder, M. Peltonen, W. Koenig, et al. Anti-inflammatory effect of lifestyle changes in the Finnish Diabetes Prevention Study Diabetologia, 52 (2009), pp. 433-442 CrossRefView Record in ScopusGoogle Scholar 81 J.H. O’Keefe, N.M. Gheewala, J.O. O’Keefe Dietary strategies for improving post-prandial glucose, lipids, inflammation, and cardiovascular health J Am Coll Cardiol, 51 (2008), pp. 249-255 ArticleDownload PDFView Record in ScopusGoogle Scholar 82 L. Monnier, E. Mas, C. Ginet, et al. Activation of oxidative stress by acute glucose fluctuations compared with sustained chronic hyperglycemia in patients with type 2 diabetes JAMA, 295 (2006), pp. 1681-1687 CrossRefView Record in ScopusGoogle Scholar 83 Reference deleted in author review. Google Scholar 84 M. Bulló, R. Casas, M.P. Portillo, et al. Dietary glycemic index/load and peripheral adipokines and inflammatory markers in elderly subjects at high cardiovascular risk Nutr Metab Cardiovasc Dis, 23 (2013), pp. 443-450 ArticleDownload PDFView Record in ScopusGoogle Scholar 85 M.L. Neuhouser, Y. Schwarz, C. Wang, et al. A low-glycemic load diet reduces serum C-reactive protein and modestly increases adiponectin in overweight and obese adults J Nutr, 142 (2012), pp. 369-374 CrossRefView Record in ScopusGoogle Scholar 86 American Heart Association Nutrition Committee, A.H. Lichtenstein, L.J. Appel, et al. Diet and lifestyle recommendations revision 2006: a scientific statement from the American Heart Association Nutrition Committee Circulation, 114 (2006), pp. 82-96 CrossRefView Record in ScopusGoogle Scholar 87 G. Egger, J. Dixon Should obesity be the main game? Or do we need an environmental makeover to combat the inflammatory and chronic disease epidemics? Obes Rev, 10 (2009), pp. 237-249 CrossRefView Record in ScopusGoogle Scholar 88 A. Costabile, A. Klinder, F. Fava, et al. Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: a double-blind, placebo-controlled, crossover study Br J Nutr, 99 (2008), pp. 110-120 View Record in ScopusGoogle Scholar 89 I. Martınez, J.M. Lattimer, K.L. Hubach, et al. Gut microbiome composition is linked to whole grain-induced immunological improvements ISME J, 7 (2013), pp. 269-280 CrossRefView Record in ScopusGoogle Scholar 90 P. Vitaglione, A. Napolitano, V. Fogliano Cereal dietary fibre: a natural functional ingredient to deliver phenolic compounds into the gut Trends Food Sci Technol, 19 (2008), pp. 451-463 ArticleDownload PDFView Record in ScopusGoogle Scholar 91 L.U. Thompson Potential health benefits of whole grains and their components Contemp Nutr, 17 (1992), pp. 1-2 View Record in ScopusGoogle Scholar 92 M. Bulló, P. Casas-Agustench, P. Amigó-Correig, et al. Inflammation, obesity and comorbidities: the role of diet Public Health Nutr, 10 (2007), pp. 1164-1172 View Record in ScopusGoogle Scholar 93 S.G. Wannamethee, G.D. Lowe, A. Rumley, et al. Associations of vitamin C status, fruit and vegetable intakes, and markers of inflammation and hemostasis Am J Clin Nutr, 83 (2006), pp. 567-574 View Record in ScopusGoogle Scholar 94 Centers for Disease Control and Prevention. State indicator report on fruits and vegetables median daily vegetable intake 2013. 2013;1–16. Google Scholar 95 Centers for Disease Control and Prevention Strategies to prevent obesity and other chronic diseases: the CDC guide to strategies to increase the consumption of fruits and vegetables U.S. Department of Health and Human Services, Atlanta (2011) Google Scholar 96 X. Zhang, X. Shu, Y. Xiang, et al. Cruciferous vegetable consumption is associated with a reduced risk of total and cardiovascular disease mortality Am J Clin Nutr, 94 (2011), pp. 240-246 CrossRefView Record in ScopusGoogle Scholar 97 A. Esmaillzadeh, M. Kimiagar, Y. Mehrabi, et al. Fruit and vegetable intakes, C-reactive protein, and the metabolic syndrome Am J Clin Nutr, 84 (2006), pp. 1489-1497 View Record in ScopusGoogle Scholar 98 L.T. Cook, G.A. O’Reilly, M.I. Goran, et al. Vegetable consumption is linked to decreased visceral and liver fat and improved insulin resistance in overweight Latino youth J Acad Nutr Diet, 114 (2014), pp. 1776-1783 ArticleDownload PDFView Record in ScopusGoogle Scholar 99 K.E. Bradbury, P.N. Appleby, T.J. Key Fruit, vegetable, and fiber intake in relation to cancer risk: findings from the European Prospective Investigation into Cancer and Nutrition (EPIC) Am J Clin Nutr, 100 (Suppl 1) (2014), pp. 394S-398S CrossRefGoogle Scholar 100 A. Nanri, D. Yoshida, T. Yamaji, et al. Dietary patterns and C-reactive protein in Japanese men and women Am J Clin Nutr, 87 (2008), pp. 1488-1496 View Record in ScopusGoogle Scholar 101 X. Gao, O.I. Bermudez, K.L. Tucker Plasma C-reactive protein and homocyestine concentrations are related to frequent fruit and vegetable intake in Hispanic and non-Hispanic white elders J Nutr, 134 (2004), pp. 913-918 View Record in ScopusGoogle Scholar 102 Reference deleted in author review. Google Scholar 103 O.K. Chum, S.J. Chung, K.J. Claycombe, et al. Serum C-reactive protein concentrations are inversely associated with dietary flavonoid intake in U.S. adults J Nutr, 138 (2008), pp. 753-760 Google Scholar 104 E.M. Holt, L.M. Steffen, A. Moran, et al. Fruit and vegetable consumption and its relation to markers of inflammation and oxidative stress in adolescents J Am Diet Assoc, 109 (2009), pp. 414-421 ArticleDownload PDFView Record in ScopusGoogle Scholar 105 Reference deleted in author review. Google Scholar 106 A.L. Macready, T.W. George, M.F. Chong, et al. Flavonoid-rich fruit and vegetables improve microvascular reactivity and inflammatory status in men at risk of cardiovascular disease-FLAVURS: a randomized controlled trial Am J Clin Nutr, 99 (2014), pp. 479-489 CrossRefView Record in ScopusGoogle Scholar 107 R.K. Johnson, L.J. Appel, M. Brands, et al. Dietary sugars intake and cardiovascular health: a scientific statement from the American Heart Association Circulation, 120 (2009), pp. 1011-1020 CrossRefView Record in ScopusGoogle Scholar 108 T.C. Campbell, T.M. Campbell The China study BenBella Books, Dallas, TX (2005) Google Scholar 109 B. Millen, A. Lichtenstein, S. Abrams, et al. Scientific report of the 2015 Dietary Guidelines Advisory Committee Dietary Guidelines Advisory Committee (2015) Google Scholar 110 L. Azadbakht, M. Kimiagar, Y. Mehrabi, et al. Soy consumption, markers of inflammation, and endothelial function: a cross-over study in postmenopausal women with the metabolic syndrome Diabetes Care, 30 (2007), pp. 967-973 CrossRefView Record in ScopusGoogle Scholar 111 H.H. Hermsdorff, Abete I. Zulet MÁ, et al. A legume-based hypocaloric diet reduces proinflammatory status and improves metabolic features in overweight/obese subjects Eur J Nutr, 50 (2011), pp. 61-69 CrossRefView Record in ScopusGoogle Scholar 112 A. Pan, Q. Sun, A.M. Bernstein, et al. Red meat consumption and mortality: results from 2 prospective cohort studies Arch Intern Med, 172 (2012), pp. 555-563 View Record in ScopusGoogle Scholar 113 L. Azadbakht, A. Esmaillzadeh Red meat intake is associated with metabolic syndrome and the plasma C-reactive protein concentration in women J Nutr, 139 (2009), pp. 335-339 View Record in ScopusGoogle Scholar 114 S.H. Ley, Q. Sun, W.C. Willett, et al. Associations between red meat intake and biomarkers of inflammation and glucose metabolism in women Am J Clin Nutr, 99 (2014), pp. 352-360 CrossRefView Record in ScopusGoogle Scholar 115 J. Montonen, H. Boeing, A. Fritsche, et al. Consumption of red meat and whole-grain bread in relation to biomarkers of obesity, inflammation, glucose metabolism and oxidative stress Eur J Nutr, 52 (2013), pp. 337-345 CrossRefView Record in ScopusGoogle Scholar 116 A. Pan, Q. Sun, A.M. Bernstein, et al., Red meat consumption and mortality: results from 2 prospective cohort studies Arch Intern Med, 172 (2012), pp. 555-563 View Record in Scopus 117 S.C. Larsson, N. Orsini Red meat and processed meat consumption and all-cause mortality: a meta-analysis Am J Epidemiol, 179 (2014), pp. 282-289 CrossRefView Record in ScopusGoogle Scholar 118 I. Abete, D. Romaguera, A.R. Vieira, et al. Association between total, processed, red and white meat consumption and all-cause, CVD and IHD mortality: a meta-analysis of cohort studies Br J Nutr, 112 (2014), pp. 762-775 CrossRefView Record in ScopusGoogle Scholar 119 F. Arya, S. Egger, D. Colquhoun, et al. Differences in postprandial inflammatory responses to a ‘modern’ v. traditional meat meal: a preliminary study Br J Nutr, 104 (2010), pp. 724-728 CrossRefView Record in ScopusGoogle Scholar 120 J.M. Hodgson, N.C. Ward, V. Burke, et al. Increased lean red meat intake does not elevate markers of oxidative stress and inflammation in humans J Nutr, 137 (2007), pp. 363-367 View Record in ScopusGoogle Scholar 121 K.J. Murphy, R.L. Thomson, A.M. Coates, et al. Effects of eating fresh lean pork on cardiometabolic health parameters Nutrients, 4 (2012), pp. 711-723 CrossRefView Record in ScopusGoogle Scholar 122 D. Li, A. Ng, N.J. Mann, et al. Contribution of meat fat to dietary arachidonic acid Lipids, 33 (1998), pp. 437-440 CrossRefView Record in ScopusGoogle Scholar 123 W. Zheng, S.A. Lee Well-done meat intake, heterocyclic amine exposure, and cancer risk Nutr Cancer, 61 (2009), pp. 437-446 CrossRefView Record in ScopusGoogle Scholar 124 P. Lopez-Legarrea, R. de la Iglesia, I. Abete, et al. The protein type within a hypocaloric diet affects obesity-related inflammation: the RESMENA project Nutrition, 30 (2014), pp. 424-429 ArticleDownload PDFView Record in ScopusGoogle Scholar 125 Labonté MÈ, P. Couture, C. Richard, et al. Impact of dairy products on biomarkers of inflammation: a systematic review of randomized controlled nutritional intervention studies Am J Clin Nutr, 97 (2013), pp. 706-717 Google Scholar 126 A. Esmaillzadeh, L. Azadbakht Dairy consumption and circulating levels of inflammatory markers among Iranian women Public Health Nutr, 13 (2010), pp. 1395-1402 CrossRefView Record in ScopusGoogle Scholar 127 D. Mozaffarian, E.B. Rimm Fish intake, contaminants, and human health: evaluating the risks and the benefits JAMA, 296 (2006), pp. 1885-1899 CrossRefView Record in ScopusGoogle Scholar 128 Environmental Defense Fund: Mercury alert: is canned tuna safe to eat?https://www.edf.org/oceans/mercury-alert-canned-tuna-safe-eat. Accessed 12/24/2016. Google Scholar 129 S. Ganjali, A. Sahebkar, E. Mahdipour, et al. Investigation of the effects of curcumin on serum cytokines in obese individuals: a randomized controlled trial Scientific World J, 2014 (2014), p. 898361 Google Scholar 130 S. Chuengsamarn, S. Rattanamongkolgul, B. Phonrat, et al. Reduction of atherogenic risk in patients with type 2 diabetes by curcuminoid extract: a randomized controlled trial J Nutr Biochem, 25 (2014), pp. 144-150 ArticleDownload PDFView Record in ScopusGoogle Scholar 131 N. Akazawa, Y. Choi, A. Miyaki, et al. Curcumin ingestion and exercise training improve vascular endothelial function in postmenopausal women Nutr Res, 32 (2012), pp. 795-799 ArticleDownload PDFView Record in ScopusGoogle Scholar 132 V. Kuptniratsaikul, P. Dajpratham, W. Taechaarpornkul, et al. Efficacy and safety of Curcuma domestica extracts compared with ibuprofen in patients with knee osteoarthritis: a multicenter study Clin Interv Aging, 9 (2014), pp. 451-458 CrossRefView Record in ScopusGoogle Scholar 133 B. Chandran, A. Goel A randomized, pilot study to assess the efficacy and safety of curcumin in patients with active rheumatoid arthritis Phytother Res, 26 (2012), pp. 1719-1725 CrossRefView Record in ScopusGoogle Scholar 134 H. Hanai, T. Iida, K. Takeuchi, et al. Curcumin maintenance therapy for ulcerative colitis: randomized, multicenter, double-blind, placebo-controlled trial Clin Gastroenterol Hepatol, 4 (2006), pp. 1502-1506 ArticleDownload PDFView Record in ScopusGoogle Scholar 135 N. Chainani-Wu, E. Madden, F. Lozada-Nur, et al. High-dose curcuminoids are efficacious in the reduction in symptoms and signs of oral lichen planus J Am Acad Dermatol, 66 (2012), pp. 752-760 ArticleDownload PDFView Record in ScopusGoogle Scholar 136 A. Di Castelnuovo, S. Costanzo, V. Bagnardi, et al. Alcohol dosing and total mortality in men and women: an updated meta-analysis of 34 prospective studies Arch Intern Med, 166 (2006), pp. 2437-2445 CrossRefView Record in ScopusGoogle Scholar 137 P.E. Ronksley, S.E. Brien, B.J. Turner, et al. Association of alcohol consumption with selected cardiovascular disease outcomes: a systematic review and meta-analysis BMJ, 342 (2011), p. d671 CrossRefView Record in ScopusGoogle Scholar 138 C. Huang, J. Zhan, Y.J. Liu, et al. Association between alcohol consumption and risk of cardiovascular disease and all-cause mortality in patients with hypertension: a meta-analysis of prospective cohort studies Mayo Clin Proc, 89 (2014), pp. 1201-1210 ArticleDownload PDFView Record in ScopusGoogle Scholar 139 A.B. Petrone, J.M. Gaziano, L. Djoussé Alcohol consumption and risk of death in male physicians with heart failure Am J Cardiol, 114 (2014), pp. 1065-1068 ArticleDownload PDFView Record in ScopusGoogle Scholar 140 R. Rodrigo, A. Miranda, L. Vergara Modulation of endogenous antioxidant system by wine polyphenols in human disease Clin Chim Acta, 412 (2011), pp. 410-424 ArticleDownload PDFView Record in ScopusGoogle Scholar 141 S. Arranz, G. Chiva-Blanch, P. Valderas-Martínez, et al. Wine, beer, alcohol and polyphenols on cardiovascular disease and cancer Nutrients, 4 (2012), pp. 759-781 CrossRefView Record in ScopusGoogle Scholar 142 R. Marfella, F. Cacciapuoti, M. Siniscalchi, et al. Effect of moderate red wine intake on cardiac prognosis after recent acute myocardial infarction of subjects with type 2 diabetes mellitus Diabet Med, 23 (2006), pp. 974-981 CrossRefView Record in ScopusGoogle Scholar 143 D.W. Droste, C. Iliescu, M. Vaillant, et al. A daily glass of red wine associated with lifestyle changes independently improves blood lipids in patients with carotid arteriosclerosis: results from a randomized controlled trial Nutr J, 12 (2013), p. 147 Google Scholar 144 M. Mueller, S. Hobiger, A. Jungbauer Anti-inflammatory activity of extracts from fruits, herbs and spices Food Chem, 122 (2010), pp. 987-996 ArticleDownload PDFView Record in ScopusGoogle Scholar 145 D.P. Rakel, A. Rindfleisch Inflammation: nutritional, botanical, and mind-body influences South Med J, 98 (2005), pp. 303-310 CrossRefView Record in ScopusGoogle Scholar 146 J. Allgrove, G. Davison Dark chocolate/cocoa polyphenols and oxidative stress Polyphenols in human health and disease, Elsevier Inc. (2014), pp. 241-251 ArticleDownload PDFGoogle Scholar 147 R. di Giuseppe, A. Di Castelnuovo, F. Centritto, et al. Regular consumption of dark chocolate is associated with low serum concentrations of C-reactive protein in a healthy Italian population J Nutr, 138 (2008), pp. 1939-1945 View Record in ScopusGoogle Scholar 148 K.S. Stote, B.A. Clevidence, J.A. Novotny, et al. Effect of cocoa and green tea on biomarkers of glucose regulation, oxidative stress, inflammation and hemostasis in obese adults at risk for insulin resistance Eur J Clin Nutr, 66 (2012), pp. 1153-1159 CrossRefView Record in ScopusGoogle Scholar 149 N. Khan, O. Khymenets, M. Urpí-Sardà, et al. Cocoa polyphenols and inflammatory markers of cardiovascular disease Nutrients, 6 (2014), pp. 844-880 CrossRefView Record in ScopusGoogle Scholar 150 O.A. Tokede, J.M. Gaziano, L. Djoussé Effects of cocoa products/dark chocolate on serum lipids: a meta-analysis Eur J Clin Nutr, 65 (2011), pp. 879-886 CrossRefView Record in ScopusGoogle Scholar 151 B.K. McFarlin, A.S. Venable, A.L. Henning, et al. Natural cocoa consumption: potential to reduce atherogenic factors? J Nutr Biochem, 26 (2015), pp. 626-632 ArticleDownload PDFView Record in ScopusGoogle Scholar 152 S. Martínez-López, B. Sarriá, J.L. Sierra-Cinos, et al. Realistic intake of a flavanol-rich soluble cocoa product increases HDL-cholesterol without inducing anthropometric changes in healthy and moderately hypercholesterolemic subjects Food Funct, 5 (2014), pp. 364-374 CrossRefView Record in ScopusGoogle Scholar 153 D. Grassi, G. Desideri, S. Necozione, et al. Cocoa consumption dose-dependently improves flow-mediated dilation and arterial stiffness decreasing blood pressure in healthy individuals J Hypertens, 33 (2015), pp. 294-303 CrossRefView Record in ScopusGoogle Scholar 154 S.G. West, M.D. McIntyre, M.J. Piotrowski, et al. Effects of dark chocolate and cocoa consumption on endothelial function and arterial stiffness in overweight adults Br J Nutr, 111 (2014), pp. 653-661 CrossRefView Record in ScopusGoogle Scholar 155 A.J. Flammer, I. Sudano, M. Wolfrum, et al. Cardiovascular effects of flavanol-rich chocolate in patients with heart failure Eur Heart J, 33 (2012), pp. 2172-2180 CrossRefView Record in ScopusGoogle Scholar 156 L. Loffredo, L. Perri, E. Catasca, et al. Dark chocolate acutely improves walking autonomy in patients with peripheral artery disease J Am Heart Assoc, 3 (4) (2014) pii: e001072 Google Scholar 157 L. Hooper, C. Kay, A. Abdelhamid, et al. Effects of chocolate, cocoa, and flavan-3-ols on cardiovascular health: a systematic review and meta-analysis of randomized trials Am J Clin Nutr, 95 (2012), pp. 740-751 CrossRefView Record in ScopusGoogle Scholar 158 C. Matsumoto, A.B. Petrone, H.D. Sesso, et al. Chocolate consumption and risk of diabetes mellitus in the Physicians’ Health Study Am J Clin Nutr, 101 (2015), pp. 362-367 CrossRefView Record in ScopusGoogle Scholar 159 B. Buijsse, E.J. Feskens, F.J. Kok, et al. Cocoa intake, blood pressure, and cardiovascular mortality: the Zutphen Elderly Study Arch Intern Med, 166 (2006), pp. 411-417 CrossRefView Record in ScopusGoogle Scholar 160 Reference deleted in author review. Google Scholar 161 Reference deleted in author review. Google Scholar 162 Y. Papanikolaou, J. Brooks, C. Reider, et al. U.S. adults are not meeting recommended levels for fish and omega-3 fatty acid intake: results of an analysis using observational data from NHANES 2003-2008 Nutr J, 13 (2014), p. 31 Google Scholar 163 Reference deleted in author review. Google Scholar 164 Reference deleted in author review. Google Scholar 165 E. Vaghef-Mehrabany, B. Alipour, A. Homayouni-Rad, et al. Probiotic supplementation improves inflammatory status in patients with rheumatoid arthritis Nutrition, 30 (2014), pp. 430-435 ArticleDownload PDFView Record in ScopusGoogle Scholar 166 M. Doménech, P. Roman, J. Lapetra, et al. Mediterranean diet reduces 24-hour ambulatory blood pressure, blood glucose, and lipids: one-year randomized, clinical trial Hypertension, 64 (2014), pp. 69-76 CrossRefView Record in ScopusGoogle Scholar 167 Reference deleted in author review. Google Scholar 168 J.S. Perona, J. Cañizares, E. Montero, et al. Virgin olive oil reduces blood pressure in hypertensive elderly subjects Clin Nutr, 23 (2004), pp. 1113-1121 ArticleDownload PDFView Record in ScopusGoogle Scholar 169 R. Moreno-Luna, R. Muñoz-Hernandez, M.L. Miranda, et al. Olive oil polyphenols decrease blood pressure and improve endothelial function in young women with mild hypertension Am J Hypertens, 25 (2012), pp. 1299-1304 View Record in ScopusGoogle Scholar 170 P. Sexton, P. Black, P. Metcalf, et al. Influence of Mediterranean diet on asthma symptoms, lung function, and systemic inflammation: a randomized controlled trial J Asthma, 50 (2013), pp. 75-81 CrossRefView Record in ScopusGoogle Scholar 171 E. Keranis, D. Makris, P. Rodopoulou, et al. Impact of dietary shift to higher-antioxidant foods in COPD: a randomised trial Eur Respir J, 36 (2010), pp. 774-780 CrossRefView Record in ScopusGoogle Scholar 172 Reference deleted in author review. Google Scholar 173 J. Salas-Salvadó, M. Bulló, N. Babio, et al. Reduction in the incidence of type 2 diabetes with the Mediterranean diet: results of the PREDIMED-Reus nutrition intervention randomized trial Diabetes Care, 34 (2011), pp. 14-19 CrossRefView Record in ScopusGoogle Scholar 174 C. Itsiopoulos, L. Brazionis, M. Kaimakamis, et al. Can the Mediterranean diet lower HbA1c in type 2 diabetes? Results from a randomized cross-over study Nutr Metab Cardiovasc Dis, 21 (2011), pp. 740-747 ArticleDownload PDFView Record in ScopusGoogle Scholar 175 K. Esposito, M.I. Maiorino, M. Ciotola, et al. Effects of a Mediterranean-style diet on the need for antihyperglycemic drug therapy in patients with newly diagnosed type 2 diabetes: a randomized trial Ann Intern Med, 151 (2009), pp. 306-314 CrossRefView Record in ScopusGoogle Scholar 176 Reference deleted in author review. Google Scholar 177 R.B. Singh, G. Dubnov, M.A. Niaz, et al. Effect of an Indo-Mediterranean diet on progression of coronary artery disease in high risk patients (Indo-Mediterranean Diet Heart Study): a randomised single-blind trial Lancet, 360 (2002), pp. 1455-1461 ArticleDownload PDFView Record in ScopusGoogle Scholar 178 Reference deleted in author review. Google Scholar 179 Reference deleted in author review. Google Scholar 180 Reference deleted in author review. Google Scholar 181 M. Ruiz-Canela, R. Estruch, D. Corella, et al. Association of Mediterranean diet with peripheral artery disease: the PREDIMED randomized trial JAMA, 311 (2014), pp. 415-417 CrossRefView Record in ScopusGoogle Scholar 182 Reference deleted in author review. Google Scholar 183 D.B. Panagiotakos, C. Chrysohoou, C. Pitsavos, et al. Association between the prevalence of obesity and adherence to the Mediterranean diet: the ATTICA study Nutrition, 22 (2006), pp. 449-456 ArticleDownload PDFView Record in ScopusGoogle Scholar 184 M. Bes-Rastrollo, A. Sánchez-Villegas, C. de la Fuente, et al. Olive oil consumption and weight change: the SUN prospective cohort study Lipids, 41 (2006), pp. 249-256 CrossRefView Record in ScopusGoogle Scholar 185 K. McManus, L. Antinoro, F. Sacks A randomized controlled trial of a moderate-fat, low-energy diet compared with a low fat, low-energy diet for weight loss in overweight adults Int J Obes Relat Metab Disord, 25 (2001), pp. 1503-1511 CrossRefView Record in ScopusGoogle Scholar 186 Reference deleted in author review. Google Scholar 187 B.C. Olendzki, T.D. Silverstein, G.M. Persuitte, et al. An anti-inflammatory diet as treatment for inflammatory bowel disease: a case series report Nutr J, 13 (2014), p. 5 Google Scholar 188 P. Nigam, S. Bhatt, A. Misra, et al. Effect of a 6-month intervention with cooking oils containing a high concentration of monounsaturated fatty acids (olive and canola oils) compared with control oil in male Asian Indians with nonalcoholic fatty liver disease Diabetes Technol Ther, 16 (2014), pp. 255-261 CrossRefView Record in ScopusGoogle Scholar 189 H.M. Parker, N.A. Johnson, C.A. Burdon, et al. Omega-3 supplementation and non-alcoholic fatty liver disease: a systematic review and meta-analysis J Hepatol, 56 (2012), pp. 944-951 ArticleDownload PDFView Record in ScopusGoogle Scholar 190 L. Schwingshackl, G. Hoffmann Adherence to Mediterranean diet and risk of cancer: a systematic review and meta-analysis of observational studies Int J Cancer, 135 (2014), pp. 1884-1897 CrossRefView Record in ScopusGoogle Scholar 191 E.H. Martínez-Lapiscina, P. Clavero, E. Toledo, et al. Mediterranean diet improves cognition: the PREDIMED-NAVARRA randomised trial J Neurol Neurosurg Psychiatry, 84 (2013), pp. 1318-1325 CrossRefView Record in ScopusGoogle Scholar 192 C. Valls-Pedret, A. Sala-Vila, M. Serra-Mir, et al. Mediterranean diet and age-related cognitive decline: a randomized clinical trial JAMA Intern Med, 175 (2015), pp. 1094-1103 CrossRefView Record in ScopusGoogle Scholar 193 C. Féart, C. Samieri, V. Rondeau, et al. Adherence to a Mediterranean diet, cognitive decline, and risk of dementia JAMA, 302 (2009), pp. 638-648 CrossRefView Record in ScopusGoogle Scholar 194 L. Shinto, J. Quinn, T. Montine, et al. A randomized placebo-controlled pilot trial of omega-3 fatty acids and alpha lipoic acid in Alzheimer’s disease J Alzheimers Dis, 38 (2014), pp. 111-120 View Record in ScopusGoogle Scholar 195 G. Grosso, A. Pajak, S. Marventano, et al. Role of omega-3 fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials PLoS One, 9 (2014), p. e96905 CrossRefGoogle Scholar 196 L. Gertsik, R.E. Poland, C. Bresee, et al. Omega-3 fatty acid augmentation of citalopram treatment for patients with major depressive disorder J Clin Psychopharmacol, 32 (2012), pp. 61-64 CrossRefView Record in ScopusGoogle Scholar 197 B. Guida, A. Napoleone, R. Trio, et al. Energy-restricted, n-3 polyunsaturated fatty acids-rich diet improves the clinical response to immuno-modulating drugs in obese patients with plaque-type psoriasis: a randomized control clinical trial Clin Nutr, 33 (2014), pp. 399-405 ArticleDownload PDFView Record in ScopusGoogle Scholar Copyright © 2018 Elsevier Inc. All rights reserved.