Take a look at the Recent articles

Circulating immune cell activation and diet: A review on human trials

Rosa Casas

Department of Internal Medicine, Hospital Clinic, Institut d'Investigació Biomèdica August Pii Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036 Barcelona, Spain (RC, AM R-L, RE).

Ciber Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain (RC, AM R-L, RE).

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

Ana María Ruiz-León

Department of Internal Medicine, Hospital Clinic, Institut d'Investigació Biomèdica August Pii Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036 Barcelona, Spain (RC, AM R-L, RE).

Ciber Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain (RC, AM R-L, RE).

Ramon Estruch

Department of Internal Medicine, Hospital Clinic, Institut d'Investigació Biomèdica August Pii Sunyer (IDIBAPS), University of Barcelona, Villarroel 170, 08036 Barcelona, Spain (RC, AM R-L, RE).

Ciber Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain (RC, AM R-L, RE).

DOI: 10.15761/JAI.1000102

Article
Article Info
Author Info
Figures & Data

Abstract

The World Health Organization (WHO) recognizes that diet plays an important role in the prevention of several low-grade inflammatory diseases such as diabetes, atherosclerosis, metabolic syndrome and obesity. All of these diseases are characterized by elevated concentrations of inflammatory markers in the systemic circulation [e.g: C-reactive protein (CRP), interleukins, fibrinogen and adhesion molecules including E-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular adhesion protein-1 (VCAM-1)].

This review focuses on the evidence obtained from epidemiological, dietary intervention and supplementation studies in humans supporting the role of monounsaturated and polyunsaturated fatty acids and other specific components of the diet in the prevention or delay of diabetes, cancer, cardiovascular disease and obesity. Thus, we provide an update of the knowledge of the relationship between diet and the modulation of immune cell activation by different dietary patterns and also highlight the importance of the overall quality and composition of the diet to protect against the previously mentioned disorders.

Key words

adhesion molecules, cytokines, dietary pattern, nutrition, inflammation metabolic syndrome

Introduction

Human health is influenced by numerous factors some of which are modifiable [1,2]. In this regard, diet is a key element because of its impact and affordability [3]. Nevertheless, although the prevalence of nutritional deficiencies has decreased, they continue to be responsible for thousands of deaths worldwide [4] as are many non-communicable diseases such as cardiovascular disease (CVD), type 2 diabetes mellitus (T2DM), and cancer [5], which have also been related to nutritional patterns and represent more than 50% of global deaths [4].

The functionality of the immune system is closely related to nutritional patterns throughout life. In healthy conditions, diet and nutrients can regulate immune activity by direct interaction with the immune cells, via receptor-mediated signalling, or indirectly, by modulation of microbiota metabolites [6]. For instance, there is growing evidence that nutritional status of the mother or food exposure during gestation may modify the probability of allergies in postnatal life [7]. In addition, breastfeeding has a key role in maturation of the immune system in newborns [8,9], ensuring the proliferation of a healthy and balanced microbiota which is necessary for correct immunological development [9]. These mechanisms are also present throughout the ageing process, albeit with adaptations, because of their association with immunosenescence [10,11].

Multiple single nutrients have shown beneficial effects on the immune system, such as vitamin A, required for maintenance of normal adaptive immunity [12] and ω-3 polyunsaturated fatty acids (PUFAs) which are related to immune response. In particular, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are involved in the regulation of the formation and action of pro-inflammatory eicosanoid, and are converted into anti-inflammatory molecules [13,14]. However, it is important consider limitations in the study of specific nutrients study since possible interactions with other diet components may be overlooked.

Overall, traditional dietary patterns have undergone changes in recent years [15]. Rapid urbanization has led to a rise in the availability of processed foods making the consumption of a Western diet increasingly more frequent. This diet is characterized by low vegetable and fruit consumption, a high presence of refined grains and high fat meat, enriched in salt, saturated fatty acids (SFA), ω-9 MUFAs (oleic acid), and is frequently associated with excess caloric intake [16]. Thus, the Western diet is low in dietary fibre [17] and high in compounds such as phosphatidylcholine and L-carnitine, which together seem to alter the microbiota and promote a state of systemic low-grade inflammation even in healthy subjects [18,19]. Rocha et al. [20] have suggested that SFA may induce inflammatory response via troll-like receptor 4 (TLR4), promoting pro-inflammatory transcript factors and activate pro-inflammatory cytokines and chemokines, which are associated with multiple chronic diseases as discussed below.

By contrast, plant-based diets like the Mediterranean diet (MeDiet) and vegetarian diets have been recommended for the maintenance of health [21]. The MeDiet, is related to anti-inflammatory processes [22–24], being composed of fruits, vegetables and the use of olive oil, and fish consumption [25]. It is rich in dietary fibre which is correlated with an abundance of faecal short chain fatty acids (SCFA), that are key to the development of healthy microbiota [11]. Other dietary patterns are also enriched in fibre including those of rural African or traditional Japanese diet as well as diets from other Asian countries. These diets frequently include large amounts of rice, beans, fermented or picked fresh foods and fish, being rich in SCFA and ω-3 fatty acids and associated with low rates of chronic inflammatory diseases [26,27].

However, protein energy malnutrition is associated with oedema, skin rash and anorexia [28], resulting in secondary immunodeficiency and vitamin A and zinc deficiencies that lead to infectious complications [29]. By contrast, some studies evaluating the effect of intermittent caloric restriction in humans without malnutrition and have reported a reduction in pro-inflammatory biomarkers [30–32].

Therefore, although activation of the immune system and the inflammatory process by diet-associated events is a complex process, there seems to be clear connection between diet, the immune system and the development of chronic inflammatory diseases.

This review provides an update of the knowledge of the mechanisms by which diet can modulate immune cell activation in chronic inflammatory diseases through the analysis of human interventional trials.

Effects of diet on chronic inflammatory diseases

The inflammatory process alters physiological responses and involves complex cell interactions and cascades of chemical mediators. One of the most characteristic effects of inflammatory activity is tissue invasion by monocytes, macrophages and lymphocytes, particularly pro-inflammatory M1 macrophages [33,34], leading to increased synthesis and secretion of pro-inflammatory molecules as well as inhibition of anti-inflammatory compounds and immune cell activation. Indeed, many of these cells or compounds and cells can be used as biomarkers of inflammatory status, including interleukins (IL-6, TNF-α, IL-18), adhesion molecules (e.g. E-selectin, intracellular adhesion molecule-1 (ICAM-1) or vascular adhesion protein-1 (VCAM-1), and C - reactive protein (CRP).

Chronic low-grade  inflammation is associated with T2DM [35], CVD [36], obesity [37], metabolic syndrome (MetS) [38,39], dementia [40], depressive disorders [41], certain types of cancer [42] and a higher risk of all-cause mortality in old age [43]. The relationship between diet and inflammatory markers has been reported in several cohort and intervention studies [44].

Obesity

Obesity can be defined as a complex disease of multifactorial causes which is due to an excessive accumulation of body fat [45]. It is characterized by the presence of macrophages infiltrated into adipose tissue (AT).  This infiltration may be due to the death of hypertrophied fat cells and/or a hypersecretion of proinflammatory cytokines by AT, including interleukin-1 (IL-1β), IL-6, IL-8, tumor necrosis factor (TNF-α), complement C3, chemoattractant molecules such as monocyte chemoattractant protein-1 (MCP-1) and macrophage migration inhibitory factor (MIF), and immune cells such as dendritic and T cells, in addition to macrophages [46]. These pro-inflammatory cytokines can also substantially affect insulin sensitivity and endothelial dysfunction, promoting an increased risk for T2DM, CVD and cancer [47-49]. Thus, the production of these molecules is directly proportional to the amount of AT, although some retrospective studies have found higher concentrations of these markers in Western diets and in those with a predominant consumption of red meat [50]. Endothelial dysfunction leads to an increase in the expression of cell adhesion molecules (CAMs) on the surface of the endothelium, increasing their interaction with circulating leukocytes [48,49]. Several studies have described a correlation between dietary patterns and inflammatory response (MCP-1 and expression of CAMs). As shown in Table 1, Ziccardi et al. [51] reported a decrease in circulating levels of proinflammatory cytokines and endothelial function in apparently health obese women with different degrees of central adiposity after one year of follow up on the MeDiet. Thompson et al. [52] allocated 90 healthy obese men and women to one of 3 energy-restricted diets for 48 weeks. Their findings showed that CRP, leptin, fasting glucose, and insulin levels significantly improved, but there were no significant differences between the experimental diets and the control diet. On the other hand, using a crossover design, Zemel et al. [53] compared the effect of 2 isoenergetic diets supplemented with smoothies made with nonfat dry milk or soy-based placebo. They found an improvement  in biomarker concentrations (TNF-α, IL-6, MCP-1 and adiponectin) in subjects consuming the dairy-based smoothies.

In another study with a 3-month follow-up in healthy obese women, it was reported that weight reduction led to a decrease in soluble adhesion molecules, suggesting a downregulation of endothelial activation [54]. On the other hand, in a study carried out in overweight subjects after weight loss, Bladbjerg and et al. [55] also reported that monounsaturated fatty acids (MUFA) and low fat diets (LFD) had similar long-term effects on inflammation and endothelial cell function. A cross-sectional study of 730 overweight women aged 43–69 years from the Nurses’ Health Study [56]  evaluated whether trans fatty acid intake could also affect biomarkers of inflammation and endothelial dysfunction including CRP, IL-6, soluble tumor necrosis factor receptor 2 (sTNFR-2), E-selectin, and sICAM-1 and sVCAM-1. It was found that trans fatty acid intake was positively related to plasma concentration of CRP (P<0.009), sTNFR-2 (P< 0.002), E-selectin (P<0.003), sICAM-1 (P< 0.007), and sVCAM-1 (P< 0.001). Another study in the same cohort [57] evaluated the effect of a prudent diet and a Western diet on CRP, IL-6, E-selectin, and sICAM-1 and sVCAM-1. The results showed a negative correlation between a healthy dietary pattern and CRP, E-selectin, VCAM-1 and ICAM-1 and a positive correlation between a Western dietary pattern and the same biomarkers of inflammation similar to the results described in another study by Esmaillzadeh et al. [58]. Diets rich in α-Linolenic acid (ALA) have also shown a greater decrease in E-selectin, VCAM-1, and ICAM-1 levels than other diets [59]. A randomised, crossover study in 11 healthy obese and overweight volunteers who consumed three high fat milkshakes rich in MUFA, SFA, or ω3-PUFA showed a higher postpandrial effect on NF-κβ than SFA while CRP was increased in the three diets, TNF-α and VCAM tended to decrease following the meals in the three diets, and ω3-PUFA enhanced NF-κβ activation compared to SFA after 6 hours of intervention [60]. It has been reported that plasma IL-6, TNF-α and VCAM-1 concentrations decreased in overweight men after a ω-6 PUFA-rich meal, while these markers ncreased after a SFA-rich meal [61]. In contrast, Manning et al. [62] showed that high-fat meals increased IL-6, independently of the type of fatty acid and had no impact on IL-8 and TNF-α concentrations.

Metabolic syndrome

Table 1 shows that  in a 12-week parallel-group study [63] in 40 obese or overweight subjects with MetS receiving an adequate-dairy diet, TNF-α, IL-6 MCP-1 and CRP concentrations significantly reduced -35%, -21%, -24% and -47%, respectively, whereas adiponectin concentrations increased by 53%. No changes were observed in subjects on a low-dairy diet. Another study [64] compared the effect of a very-low-carbohydrate diet (LC) and an isocaloric high-carbohydrate (HC) in men and women with overweight or obesity on markers of endothelial function and CVD risk in the short-term. The results showed a decrease in endothelial markers, E- and P- selectin and ICAM-1 (P< 0.001), independently of the diet and observed a small, albeit significant, increase in VCAM-1. However, a similar study with a LFD and a LC diet showed an improvement in adiponectin and E-Selectin levels in both groups (P < 0.01 for time) in the long-term [65], although ICAM-1 only decreased with the LC diet and no changes were observed in VCAM-1. In addition, in overweight or obese men with MetS, Roberts and et al. [66] showed a reduction in oxidative stress and inflammatory parameters or markers related to vulnerability plaque such as MMP-9, after one month of intervention with diet and physical exercise, independently of weight loss since the magnitude of weight loss was minimal. A parallel-group intervention study [67] reported no changes in CRP, IL-6 and TNF-α concentrations with either of two diets (a diet based on the consumption of 3–5 portions of dairy products daily and control group) after 6 months of intervention in 121 overweight subjects with MetS. On the other hand, a randomised, single-blind trial assessed the effect of a MeDiet on endothelial function and vascular inflammatory markers in 180 patients with the MetS after two years of follow-up [68]. The findings showed that serum concentrations of CRP, IL-6, IL-7, and IL-18 had significantly reduced in patients consuming the MeDiet and the endothelial function score improved in the intervention group but remained stable in the control group. Finally, the CRP levels of teenagers with MetS reduced after six weeks of a Dietary Approaches to Stop Hypertension (DASH) diet, independently of weight loss or the lipid profile. Nonetheless, there was no significant modification in TNF-α, IL-2, IL-6 or adiponectin levels in this population [69].

Type 2 diabetes

As shown in table 1, several observational [70] and interventional [71] studies have reported that high intake of refined grains causes rapid swings in blood glucose and insulin concentrations, may increase hunger, and elevate free fatty acid levels thereby promoting hyperglycaemia and increasing circulating levels of free radicals and proinflammatory cytokines such as IL-6, IL-18, and TNF-α [72]. On the other hand, Shai and et al. [73] compared the effectiveness and safety of three weight-loss diets on healthy obese individuals over 2 years of intervention. The diets were: a calorie- restricted LFD, a calorie- restricted MeDiet or a non– calorie- restricted LC diet,. After a weight loss of 4.7 kg in the LC diet, 4.4 kg for the MeDiet and 2.9 kg in the LFD, CRP levels decreased in both the LC diet and the MeDiet. Moreover, the results of weight loss due to laparoscopic gastric banding indicate that weight loss is more important than glycaemic control in regulating circulating levels of ICAM-1, endothelin-1, E-selectin in morbidly obese subjects [74]. In the Nurses’ Health Study [75] including 902 diabetic women significant decreases in CRP (P for trend = 0.03 and 0.007, respectively) and TNF-R2 (P for trend=0.017 and 0.06, respectively) were observed after whole grain and bran intake. While a significant decrease in IL-18 and a significant increase in adiponectin concentrations were observed in an interventional study with coffee carried out in 47 subjects under the age of 65 years who did not have T2DM but had an elevated risk for the development of T2DM,  [76], CRP, leptin, Serum amyloid A (SAA), IL-6, MIF and IL-1ra concentrations did not change. A meta-analysis of 20 cohort studies found that moderate alcohol consumption also protects against diabetes [77]. Estruch et al. [78] described the different effects of red wine and polyphenol-free gin consumption in a prospective randomised crossover study on 40 healthy men (mean age, 37.6 years) who consumed 30 g ethanol per day as either wine or gin for 28 days. Both wine and gin showed anti-inflammatory effects by reducing plasma fibrinogen and IL-1α levels. However, wine showed an additional effect of decreasing CRP (-21%), VCAM-1 (-17%) and ICAM-1 (-9%) levels, as well as monocyte and endothelial adhesion molecules. Finally, Ceriello et al. [79] suggested that the MeDiet supplemented with olive oil, prevents acute hyperglycaemia effects on endothelial function, inflammation and oxidative stress, and improves the action of GLP-1 in the management of T2DM.

Cardiovascular disease

According to several cross-sectional studies, the consumption of dairy products is inversely associated with low-grade systemic inflammation [24, 80]. As shown in Table 1, the ATTICA study [80] including 3,042 subjects without CVD, found that TNF-α (-20%), IL-6 (-9%) and CRP (-29%) concentrations were lower in subjects consuming >2 servings of dairy products per day than in individuals consuming ≤1 serving per day. Salas-Salvadó et al. [24] found that lower IL-6 concentrations were associated with a higher consumption of fruits and cereals. In addition, a higher consumption of nuts and extra virgin olive oil (EVOO) were correlated with lower ICAM-1 and VCAM-1 concentrations, respectively. The Multi-Ethnic Study of Atherosclerosis (MESA) reported that frequent nut and seed consumption was associated with lower levels of inflammatory markers such as CRP and IL-6 [81]. In the large intervention PREDIMED (Prevención con Dieta Mediterránea) trial including 7,447 subjects (50% men 55–80 years and 50% women, 60–80 years) with diabetes or who met at least three or more other CVD risk factors it was found that a MeDiet rich in EVOO or nuts reduced the risk of CVD by 30% after a mean follow-up of nearly 5 years compared to a LFD [82]. This study also demonstrated that higher adherence to the MeDiet may also exert a modulatory effect on the expression of genes related to plaque stability, such as MMP-9, even after a short-term in an elderly high-risk population [83]. After a 3- and 12-month and 3- and 5-year intervention, the MeDiet also demonstrated anti-inflammatory effects on the expression of adhesion molecules in leukocyte cell membranes (T-Lymphocytes and monocytes) and circulating endothelial adhesion molecules (VCAM-1, ICAM-1, E- and P-Selectin), cytokines (IL-1, TNF-α, IL-6, CRP, TNFR-60 and TNFR-80 etc.) and molecules related to atherosclerotic plaque instability (IL-18, MMP-9), all of which are related to the atherosclerotic process [84-88]. In particular, antioxidants and polyphenols have been shown to exert a protective role against ischaemic CVD mainly due to their anti-inflammatory properties [89,90]. Monagas et al. [91] studied the effect of a 4-week randomised crossover trial with 40 g cocoa powder in skimmed milk daily vs. skimmed milk alone in 42 older subjects with high cardiovascular risk on plasma concentrations of sICAM-1 and sP-selectin and on monocyte expression of VLA-4, CD49d, CD36 and CD40 and found a significant reduction of all these inflammatory factors after the cocoa powder intervention. Also a recent critical review [92] including 33 randomised clinical trials concluded that acute cocoa consumption may reduce inflammation by a reduced activation of monocytes and neutrophils, a decrease in the expression of adhesion molecules (VCAM-1, ICAM-1, E- and P-selectin) and 4-series leukotrienes in serum as well as a decrease in the activation of NF- κβ in peripheral leukocytes. Furthermore, Chiva-Blanch et al. [93] also reported that red wine and gin interventions increased anti-inflammatory IL-10 and decreased IL-16, while red wine but not gin decreased IL-6 plasma concentrations in high cardiovascular risk patients. Valderas-Martinez et al. [94] carried out a randomised, controlled feeding intervention trial to evaluate the postprandial effects of a single dose of raw tomato (RT), tomato sauce cooked without oil (TS) and tomato sauce cooked with refined olive oil (TSOO) on markers of inflammation and CVD. They found that tomato intake decreased some inflammatory biomarker concentrations such as LFA-1, IL-6, IL-18, MCP-1 and VCAM-1, and increased plasma IL-10 concentrations. Moreover, their findings demonstrated that tomato, especially that cooked and enriched with oil, enhanced the effects of tomato intake on the cardiovascular system, since the effects of TSOO on plasma inflammatory biomarker concentrations were greater than those of RT and TS.

Cancer, dementia and other pathologies

In a tranversal study, Souza et al. [95] investigated the relationship between the anthropometric profile [body mass index (BMI) and waist-to-hip ratio], food intake frequency, the lipid profile and fasting glycaemia and serum adipokine (adiponectin and PAI-1) and adhesion molecules levels (ICAM-1 and VCAM-1) in a subgroup of women derived from a cohort of 10,000 women. A total of 145 women over 40 years of age participated in this study. No association was found between dietary intake and the amount of calories/day ingested and serum adipokine and adhesion molecule levels. A linear correlation was found between serum VCAM-1 levels and the BMI. A cross-sectional study examined fibre intake and 17 cytokines and chemokines in 88 healthy participants in the EPIC-Italy study and observed an association between cereal fibre and decreased levels of predominantly pro-inflammatory cytokines (IL-1β, IL-4, IL-5, IL-6, IL-13, and TNF-α), although there were no associations with fruit and vegetable fibre [96]. Recently, Schmandt et al. [97] reported that weight loss (a 10%) reduces colorectal inflammation and the risk of colorectal cancer. Weight loss was associated with a significant decrease in a variety of inflammatory cytokines including TNF-α, IL-6, IL-8, and MCP-1, as well as downregulation of markers of proinflammatory pathways, prostaglandin metabolism and transcription factors.

On the other hand, the consumption of nuts has also been associated with decreased plasma concentrations of VCAM-1, ICAM-1 and sE-selectin in a crossover study in 12 healthy subjects and 12 patients with hypercholesterolemia after intaking 2 high-fat meals including 25 g of olive oil or 40 g of walnuts [98]. In addition, an interventional study has also shown significant reductions in CRP, VCAM-1 and E-selectin  after dietary supplementation with α-linolenic acid (ALA) (6.5% from total energy) in 23 hypercholesterolemic patients (20 men, 26-60 years, and 3 women, 55-65 years) [99]. Finally, dietary supplementation with ALA (15 ml of linseed oil/day) for 3 months in 50 dyslipidemic patients with a mean age of 41 years, significantly decreased CRP, SAA and IL-6 concentrations by 38%, 23%  and10%, respectively. Contrarily to ω-3, ω-6 PUFA (linoleic acid) has shown to exert an inflammatory effect [100].

Brymora et al. [101] reported that a low-fructose diet in subjects (n=28, age 59 ± 15 years, 17 males/11 females) with chronic kidney disease can reduce inflammation (CRP and ICAM-1), and may also have potential benefits on blood pressure after 6 weeks of follow-up. In a recent comparative study, Gasbarrini et al. [102] evaluated innate and adaptive immunity expression in celiac disease and non-celiac gluten sensitivity. Their results demonstrated that subjects with celiac disease had a higher presence of IL-6 and IL-21 and adaptive immunity markers, while subjects with non-celiac gluten sensitivity showed an increase in the expression of TLR-2, an innate immunity marker, compared to patients with celiac disease.

Conclusion

This review demonstrates that a Mediterranean-style diet rich in whole grains, fruits, vegetables, legumes, walnuts, and olive oil might be effective in reducing both the prevalence of the most frequent chronic diseases such as T2DM, CVD, obesity or MetS compared to Western dietary patterns mainly characterized by a high intake of SFA. Diet, weight loss by hypocaloric diets with different macronutrient distribution or by surgical intervention, regular practice of physical activity and healthy lifestyle seem to be associated with a reduction of the low-grade inflammatory state linked to all these diseases. Benefits in antiinfllammatory status are associated with a higher consumption of dietary fibre, whole grain, ω-3 fatty acids and dairy products, among others. One of the mechanisms responsible for these protective effects is the reduction of cell adhesion molecules (VCAM-1, ICAM-1, E-and P-Selectin), chemokines, interleukins and other inflammatory biomarkers after the adoption of a healthy dietary pattern.

Thus, polices or nutritional recommendations focused on these healthy dietary patterns and healthy foods should be promoted by governments and scientific societies as tools for the prevention of CVD, diabetes, obesity, cancer or MetS, among other diseases.

 

Conflict of interest

The authors have no conflict of interest.

Acknowledgements

This work has been partially supported by  PIE14/00045 from the Instituto de Salud Carlos III, Spain.

Author contributions

Casas R and Ruiz-León AM substantially contributed to the conception and design and the drafting of the article; Estruch R critically revised the article for important intellectual content; and Casas R, Ruiz-León AM and Estruch R approved the final version of the manuscript for publication.

References

  1. Matheson GO, Klügl M, Engebretsen L, Bendiksen F, Blair SN, et al. (2013) Prevention and management of non-communicable disease: the IOC consensus statement, Lausanne. Br J Sports Med 47: 1003-1011. [Crossref]
  2. Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, et al. (2012) A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380: 2224-2260. [Crossref]
  3. Byrne DW, Rolando LA, Aliyu MH, McGown PW, Connor LR, et al. (2016) Modifiable Healthy Lifestyle Behaviors: 10-Year Health Outcomes From a Health Promotion Program. Am J Prev Med 51: 1027-1037. [Crossref]
  4. GBD 2015 Mortality and Causes of Death Collaborators (2016) Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980-2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388: 1459-1544. [Crossref]
  5. Schmidhuber J, Shetty P (2011) Nutrition, Lifestyle, Obesity and Chronic Disease. Expert Paper No. 2011/3.
  6. Zmora N, Bashiardes S, Levy M, Elinav E (2017) The Role of the Immune System in Metabolic Health and Disease. Cell Metab 25: 506-521. [Crossref]
  7. Lee SE, Kim H (2016) Update on Early Nutrition and Food Allergy in Children. Yonsei Med J 57: 542-548. [Crossref]
  8. Turfkruyer M, Verhasselt V (2015) Breast milk and its impact on maturation of the neonatal immune system. Curr Opin Infect Dis 28: 199-206. [Crossref]
  9. Walker WA, Iyengar RS (2015) Breast milk, microbiota, and intestinal immune homeostasis. Pediatr Res 77: 220-228. [Crossref]
  10. Yaqoob P (2016) Ageing alters the impact of nutrition on immune function. Proc Nutr Soc. [Crossref]
  11. Clements SJ, Carding SR, (2016) Diet, the intestinal microbiota and immune health in ageing. Crit Rev Food Sci Nutr. [Crossref]
  12. Bono MR, Tejon G, Flores-Santibañez F, Fernandez D, Rosemblatt M, et al. (2016) Retinoic Acid as a Modulator of T Cell Immunity. Nutrients 8. [Crossref]
  13. Arita M (2016) Eosinophil polyunsaturated fatty acid metabolism and its potential control of inflammation and allergy. Allergol Int 65: S2-5. [Crossref]
  14. Simopoulos AP (2002) Omega-3 Fatty Acids in Inflammation and Autoimmune Diseases. J Am Coll Nutr 21: 495–505.
  15. Zhang R, Wang Z, Fei Y, Zhou B, Zheng S, et al. (2015) The Difference in Nutrient Intakes between Chinese and Mediterranean, Japanese and American Diets. Nutrients 7: 4661-4688. [Crossref]
  16. Bédard A, Garcia-Aymerich J, Sanchez M, Le Moual N, Clavel-Chapelon F, et al. (2015) Confirmatory Factor Analysis Compared with Principal Component Analysis to Derive Dietary Patterns: A Longitudinal Study in Adult Women. J Nutr 145: 1559-1568. [Crossref]
  17. Maslowski KM, Mackay CR (2011) Diet, gut microbiota and immune responses. Nat Immunol 12: 5-9. [Crossref]
  18. Fogarty CL, Nieminen JK, Peräneva L, Lassenius MI, Ahola AJ, et al. (2015) High-fat meals induce systemic cytokine release without evidence of endotoxemia-mediated cytokine production from circulating monocytes or myeloid dendritic cells. Acta Diabetol 52: 315-322. [Crossref]
  19. De Lorenzo A, Bernardini S, Gualtieri P, Cabibbo A, Perrone MA, et al. (2017) Mediterranean meal versus Western meal effects on postprandial ox-LDL, oxidative and inflammatory gene expression in healthy subjects: a randomized controlled trial for nutrigenomic approach in cardiometabolic risk. Acta Diabetol 54: 141-149. [Crossref]
  20. Rocha DM, Caldas AP, Oliveira LL, Bressan J, Hermsdorff HH (2016) Saturated fatty acids trigger TLR4-mediated inflammatory response. Atherosclerosis 244: 211-215. [Crossref]
  21. 2015 Dietary Guidelines Advisory Committee Membership.
  22. Neale EP, Batterham MJ, Tapsell LC (2016) Consumption of a healthy dietary pattern results in significant reductions in C-reactive protein levels in adults: a meta-analysis. Nutr Res 36: 391-401. [Crossref]
  23. Tresserra-Rimbau A, Guasch-Ferré M, Salas-Salvadó J, Toledo E, Corella D, et al. (2016) Intake of Total Polyphenols and Some Classes of Polyphenols Is Inversely Associated with Diabetes in Elderly People at High Cardiovascular Disease Risk. J Nutr.
  24. Salas-Salvadó J, Garcia-Arellano A, Estruch R, Marquez-Sandoval F, Corella D, et al. (2008) Components of the Mediterranean-type food pattern and serum inflammatory markers among patients at high risk for cardiovascular disease. Eur J Clin Nutr 62: 651-659. [Crossref]
  25. Martínez-González MA, Zazpe I, Razquin C, Sánchez-Tainta A, Corella D, Salas-Salvadó J, et al. (2014) Empirically-derived food patterns and the risk of total mortality and cardiovascular events in the PREDIMED study. Clin Nutr 34: 859-67. [Crossref]
  26. O'Keefe SJ, Li JV, Lahti L, Ou J, Carbonero F, et al. (2015) Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun 6: 6342. [Crossref]
  27. Yang Y, Hu XM, Chen T-J, Bai M-J (2016) Rural-Urban Differences of Dietary Patterns, Overweight, and Bone Mineral Status in Chinese Students. Nutrients 8: 537. [Crossref]
  28. Tilg H, Moschen AR (2015) Food, immunity, and the microbiome. Gastroenterology 148: 1107-1119. [Crossref]
  29. Albers R, Bourdet-Sicard R, Braun D, Calder PC, Herz U, et al. (2013) Monitoring immune modulation by nutrition in the general population: identifying and substantiating effects on human health. Br J Nutr 110: S1-30. [Crossref]
  30. Johnson JB, Summer W, Cutler RG, Martin B, Hyun DH, et al. (2007) Alternate day calorie restriction improves clinical findings and reduces markers of oxidative stress and inflammation in overweight adults with moderate asthma. Free Radic Biol Med 42: 665-674. [Crossref]
  31. Testa G, Biasi F, Poli G, Chiarpotto E (2014) Calorie restriction and dietary restriction mimetics: a strategy for improving healthy aging and longevity. Curr Pharm Des 20: 2950-2977. [Crossref]
  32. Brandhorst S, Choi IY, Wei M, Cheng CW, Sedrakyan S, et al. (2015) A Periodic Diet that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cognitive Performance, and Healthspan. Cell Metab 22: 86-99. [Crossref]
  33. Lumeng CN, Bodz2021 Copyright OAT. All rights reservces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175-184. [Crossref]
  34. Chatzigeorgiou A, Chavakis T, et al. (2016) Immune Cells and Metabolism. Handb Exp Pharmacol 233: 221-249. [Crossref]
  35. Donath MY, Shoelson SE (2011) Type 2 diabetes as an inflammatory disease. Nat Rev Immunol 11: 98-107. [Crossref]
  36. Hansson GK, Hermansson A (2011) The immune system in atherosclerosis. Nat Immunol 12: 204-212. [Crossref]
  37. Adrielle Lima Vieira R, Nascimento de Freitas R, Volp AC (2014) Adhesion molecules and chemokines; relation to anthropometric, body composition, biochemical and dietary variables. Nutr Hosp 30: 223-236.
  38. Tamakoshi K, Yatsuya H, Kondo T, Hori Y, Ishikawa M (2003) The metabolic syndrome is associated with elevated circulating C-reactive protein in healthy reference range, a systemic low-grade inflammatory state. Int J Obes Relat Metab Disord 27: 443-449. [Crossref]
  39. Ford ES (2003) The metabolic syndrome and C-reactive protein, fibrinogen, and leukocyte count: findings from the Third National Health and Nutrition Examination Survey. Atherosclerosis 168: 351-358. [Crossref]
  40. Xu G, Zhou Z, Zhu W, Fan X, Liu X (2009) Plasma C-reactive protein is related to cognitive deterioration and dementia in patients with mild cognitive impairment. J Neurol Sci 284: 77-80. [Crossref]
  41. Howren MB, Lamkin DM, Suls J (2009) Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med 71: 171-186. [Crossref]
  42. Khandekar MJ, Cohen P, Spiegelman BM (2011) Molecular mechanisms of cancer development in obesity. Nat Rev Cancer 11: 886-895. [Crossref]
  43. Lee JK, Bettencourt R, Brenner D, Le TA, Barrett-Connor E, et al. (2012) Association between serum interleukin-6 concentrations and mortality in older adults: the Rancho Bernardo study. PLoS ONE 7:e34218.
  44. Minihane AM, Vinoy S, Russell WR, Baka A, Roche HM, et al. (2015). Low-grade inflammation, diet composition and health: current research evidence and its translation. Br J Nutr 114: 999-1012. [Crossref]
  45. Guh DP, Zhang W, Bansback N, Amarsi Z, Birmingham CL, et al. (2009) The incidence of co-morbidities related to obesity and overweight: a systematic review and meta-analysis. BMC Public Health 9: 88. [Crossref]
  46. Volp ACP, Barbosa KBF, Bressan J (2012) Triacylglycerols and body fat mass are possible independent predictors of C3 in apparently healthy young brazilian adults. Nutrition 28: 544-550.
  47. Berg AH, Scherer PE (2005) Adipose tissue, inflammation, and cardiovascular disease. Circ Res 96: 939-949. [Crossref]
  48. Gomes F, Telo DF, Souza HP, Nicolau JC, Halpern A, et al. (2010) [Obesity and coronary artery disease: role of vascular inflammation]. Arq Bras Cardiol 94: 255-261, 273-9, 260-6. [Crossref]
  49. Gustafson B1 (2010) Adipose tissue, inflammation and atherosclerosis. J Atheroscler Thromb 17: 332-341. [Crossref]
  50. Barbaresko J, Koch M, Schulze M, Nöthlings U (2013) Dietary pattern analysis and biomarkers of low- grade inflammation: a systematic literature review. Nutrition Reviews 71: 511-527. [Crossref]
  51. Ziccardi P, Nappo F, Giugliano G, Esposito K, Marfella R, et al. (2002) Reduction of inflammatory cytokine concentrations and improvement of endothelial functions in obese women after weight loss over one year. Circulation 105: 804-809. [Crossref]
  52. Thompson WG, Rostad Holdman N, Janzow DJ, Slezak JM, Morris KL, et al. (2005) Effect of energy-reduced diets high in dairy products and fiber on weight loss in obese adults. Obes Res 13: 1344-1353. [Crossref]
  53. Zemel MB, Sun X, Sobhani T, Wilson B (2010) Effects of dairy compared with soy on oxidative and inflammatory stress in overweight and obese subjects. Am J Clin Nutr 91: 16-22. [Crossref]
  54. Ito H, Ohshima A, Inoue M, Ohto N, Nakasuga K, et al. (2002) Weight reduction decreases soluble cellular adhesion molecules in obese women. Clin Exp Pharmacol Physiol 29: 399-404.
  55. Bladbjerg EM, Larsen TM, Due A, Stender S, Astrup A, et al. (2011) Effects on markers of inflammation and endothelial cell function of three ad libitum diets differing in type and amount of fat and carbohydrate: a 6-month randomised study in obese individuals. Br J Nutr 106: 123-129. [Crossref]
  56. Lopez-Garcia E, Schulze MB, Meigs JB, Manson JE, Rifai N, et al. (2005) Consumption of trans fatty acids is related to plasma biomarkers of inflammation and endothelial dysfunction. J Nutr 135: 562-566. [Crossref]
  57. Lopez-Garcia E (2004) Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr 80: 1029-1035. [Crossref]
  58. Esmaillzadeh A, Kimiagar M, Mehrabi Y, Azadbakht L, Hu FB, et al. (2007) Dietary patterns and markers of systemic inflammation among Iranian women. J Nutr 137: 992-998. [Crossref]
  59. Zhao G, Etherton TD, Martin KR, West SG, Gillies PJ, et al. (2004) Dietary alpha-linolenic acid reduces inflammatory and lipid cardiovascular risk factors in hypercholesterolemic men and women. J Nutr 134: 2991-2997. [Crossref]
  60. Peairs AD, Rankin JW, Lee YW (2011) Effects of acute ingestion of different fats on oxidative stress and inflammation in overweight and obese adults. Nutr J 10: 122. [Crossref]
  61. Masson CJ & Mensink RP (2011) Exchanging saturated fatty acids for (n-6) polyunsaturated fatty acids in a mixed meal may decrease postprandial lipemia and markers of inflammation and endothelial activity in overweight men. J Nutr 141: 816–821.
  62. Manning PJ, Sutherland WH, McGrath MM, de Jong SA, Walker RJ, et al. (2008) Postprandial cytokine concentrations and meal composition in obese and lean women. Obesity (Silver Spring) 16: 2046-2052. [Crossref]
  63. Stancliffe RA, Thorpe T, Zemel MB (2011) Dairy attentuates oxidative and inflammatory stress in metabolic syndrome. Am J Clin Nutr 94: 422-430. [Crossref]
  64. Keogh JB, Brinkworth GD, Noakes M, Belobrajdic DP, Buckley JD, et al. (2008) Effects of weight loss from a verylow-carbohydrate diet on endothelial function and markers of cardiovascular disease risk in subjects with abdominal obesity. Am J Clin Nutr 87: 567-576. [Crossref]
  65. Wycherley TP, Brinkworth GD, Keogh JB, Noakes M, Buckley JD, et al. (2010) Long-term effects of weight loss with a very low carbohydrate and low fat diet on vascular function in overweight and obese patients. J Intern Med 267: 452-461. [Crossref]
  66. Roberts CK, Won D, Pruthi S, Kurtovic S, Sindhu RK, et al. (2006) Effect of a short-term diet and exercise intervention on oxidative stress, inflammation, MMP-9, and monocyte chemotactic activity in men with metabolic syndrome factors. J Appl Physiol 100: 1657-1665.
  67. Wennersberg MH, Smedman A, Turpeinen AM, Retterstøl K, Tengblad S, et al. (2009) Dairy products and metabolic effects in overweight men and women: results from a 6-mo intervention study. Am J Clin Nutr 90: 960-968. [Crossref]
  68. Esposito K, Marfella R, Ciotola M, Di Palo C, Giugliano F, et al. (2004) Effect of a mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA 292: 1440-1446. [Crossref]
  69. Saneei P, Hashemipour M, Kelishadi R, Esmaillzadeh A (2014) The Dietary Approaches to Stop Hypertension (DASH) diet affects inflammation in childhood metabolic syndrome: a randomized cross-over clinical trial. Ann Nutr Metab 64: 20-27. [Crossref]
  70. Festa A, D’Agostino R Jr., Tracy RP, Haffner SM (2002) C-reactive protein is more strongly related to post-glucose load glucose than to fasting glucose in nondiabetic subjects; the Insulin Resistance Atherosclerotic Study. Diabetic Med 19: 939-943.
  71. Esposito K, Nappo F, Giugliano F, Di Palo C, Ciotola M, et al. (2003) Meal modulation of circulating interleukin 18 and adiponectin concentrations in healthy subjects and in patients with type 2 diabetes mellitus. Am J Clin Nutr 78: 1135-1140. [Crossref]
  72. Esposito K, Nappo F, Marfella R, Giugliano G, Giugliano F, et al. (2002) Inflammatory cytokine concentrations are acutely increased by hyperglycemia in humans: role of oxidative stress. Circulation 106: 2067-2072.
  73. Shai I, Schwarzfuchs D, Henkin Y, Shahar DR, Witkow S, et al. (2008) Weight loss with a low-carbohydrate, Mediterranean, or low-fat diet. N Engl J Med 359: 229-241. [Crossref]
  74. Pontiroli AE, Pizzocri P, Koprivec D, Vedani P, Marchi M, et al. (2004) Body weight and glucose metabolism have a different effect on circulating levels of ICAM-1, E-selectin, and endothelin-1 in humans. Eur J Endocrinol 150: 195-200. [Crossref]
  75. Qi L, van Dam RM, Liu S, Franz M, Mantzoros C, et al. (2006) Whole-grain, bran, and cereal fiber intakes and markers of systemic inflammation in diabetic women. Diabetes Care 29: 207-211. [Crossref]
  76. Kempf K, Herder C, Erlund I, Kolb H, Martin S, et al. (2010) Effects of coffee consumption on subclinical inflammation and other risk factors for type 2 diabetes: a clinical trial. Am J Clin Nutr 91: 950-957. [Crossref]
  77. Baliunas DO, Taylor BJ, Irving H, Roerecke M, Patra J, et al. (2009) Alcohol as a risk factor for type 2 diabetes: A systematic review and meta-analysis. Diabetes Care 32: 2123-2132. [Crossref]
  78. Estruch R, Sacanella E, Badia E, Antúnez E, Nicolás JM, et al. (2004) Different effects of red wine and gin consumption on inflammatory biomarkers of atherosclerosis: a prospective randomized crossover trial. Effects of wine on inflammatory markers. Atherosclerosis 175: 117-123. [Crossref]
  79. Ceriello A, Esposito K, La Sala L, Pujadas G, De Nigris V, Testa R, et al. (2014) The protective effect of the Mediterranean diet on endothelial resistance to GLP-1 in type 2 diabetes: a preliminary report. Cardiovasc Diabetol 13: 140. [Crossref]
  80. Panagiotakos DB, Pitsavos CH, Zampelas AD, Chrysohoou CA, Stefanadis CI (2010) Dairy products consumption is associated with decreased levels of inflammatory markers related to cardiovascular disease in apparently healthy adults: the ATTICA study. J Am Coll Nutr 29: 357-364. [Crossref]
  81. Jiang R, Jacobs DR Jr, Mayer-Davis E, Szklo M, Herrington D, et al. (2006) Nut and seed consumption and inflammatory markers in the multi-ethnic study of atherosclerosis. Am J Epidemiol 163: 222-231. [Crossref]
  82. Estruch R, Ros E, Salas-Salvadó J, Covas MI, Corella D, et al. (2013) Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med 368: 1279-1290. [Crossref]
  83. Camargo A, Delgado-Lista J, Garcia-Rios A, Cruz-Teno C, Yubero-Serrano EM, et al. (2012) Expression of proinflammatory; proatherogenic genes is reduced by the Mediterranean diet in elderly people. Br J Nutr 108: 500-508. [Crossref]
  84. Estruch R, Martínez-González MA, Corella D, Salas-Salvadó J, Ruiz-Gutiérrez V, et al. (2006) Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 145: 1-11. [Crossref]
  85. Urpi-Sarda M, Casas R, Chiva-Blanch G, Romero-Mamani ES, Valderas-Martínez P, et al. (2012). The Mediterranean diet pattern and its main components are associated with lower plasma concentrations of tumor necrosis factor receptor 60 in patients at high risk for cardiovascular disease. J Nutr 142: 1019-1025. [Crossref]
  86. Casas R, Sacanella E, Urpí-Sardà M, Chiva-Blanch G, Ros E, et al. (2014) The effects of the mediterranean diet on biomarkers of vascular wall inflammation and plaque vulnerability in subjects with high risk for cardiovascular disease. A randomized trial. PLoS One 9: e100084.
  87. Mena MP, Sacanella E, Vazquez-Agell M, Morales M, Fitó M, et al. (2009) Inhibition of circulating immune cell activation: a molecular antiinflammatory effect of the Mediterranean diet. Am J Clin Nutr 89: 248-256. [Crossref]
  88. Casas R, Sacanella E, Urpí-Sardà M, Corella D, Castañer O, et al. (2016). Long-Term Immunomodulatory Effects of a Mediterranean Diet in Adults at High Risk of Cardiovascular Disease in the PREvención con DIeta MEDiterránea (PREDIMED) Randomized Controlled Trial. J Nutr 146: 1684-1693.
  89. Medina-Remón A, Casas R, Tressserra-Rimbau A, Ros E, Martínez-González MA, et al. (2017) Polyphenol intake from a Mediterranean diet decreases inflammatory biomarkers related to atherosclerosis: a substudy of the PREDIMED trial. Br J Clin Pharmacol 83: 114-128.
  90. Kang JX, Leaf A (2000) Prevention of fatal cardiac arrhythmias by polyunsaturated fatty acids. Am J Clin Nutr 71: 202S-207S. [Crossref]
  91. Monagas M, Khan N, Andres-Lacueva C, Casas R, Urpí-Sardà M, et al. (2009) Effect of cocoa powder on the modulation of inflammatory biomarkers in patients at high risk of cardiovascular disease. Am J Clin Nutr 90: 1144-1150. [Crossref]
  92. Ellinger S, Stehle P (2016) Impact of Cocoa Consumption on Inflammation Processes-A Critical Review of Randomized Controlled Trials. Nutrients 8: pii: E321. [Crossref]
  93. Chiva-Blanch G, Urpi-Sarda M, Llorach R, Rotches-Ribalta M, Guillén M, et al. (2012) Differential effects of polyphenols and alcohol of red wine on the expression of adhesionmolecules and inflammatory cytokines related to atherosclerosis: a randomized clinical trial. Am J Clin Nutr 95: 326-334. [Crossref]
  94. Valderas-Martinez P, Chiva-Blanch G, Casas R, Arranz S, Martínez-Huélamo M, et al. (2016) Tomato Sauce Enriched with Olive Oil Exerts Greater Effects on Cardiovascular Disease Risk Factors than Raw Tomato and Tomato Sauce: A Randomized Trial. Nutrients 8: 170. [Crossref]
  95. Isoppo de Souza C, Rosa DD, Ettrich B, Cibeira GH, Giacomazzi J, et al. (2012) Association of adipokines and adhesion molecules with indicators of obesity in women undergoing mammography screening. Nutr Metab (Lond) 9: 97.
  96. Chuang SC, Vermeulen R, Sharabiani MT, Sacerdote C, Fatemeh SH, et al. (2011) The intake of grain fibers modulates cytokine levels in blood. Biomarkers 16: 504-510. [Crossref]
  97. Schmandt RE, Iglesias DA, Co NN, Lu KH (2011) Understanding obesity and endometrial cancer risk: opportunities for prevention. Am J Obstet Gynecol 205: 518-525. [Crossref]
  98. Cortés B, Núñez I, Cofán M, Gilabert R, Pérez-Heras A, et al. (2006) Acute effects of high-fat meals enriched with walnuts or olive oil on postprandial endothelial function. J Am Coll Cardiol 48: 1666-1671. [Crossref]
  99. Zhao G, Etherton TD, Martin KR, West SG, Gillies PJ, et al. (2004) Dietary alpha-linolenic acid reduces inflammatory and lipid cardiovascular risk factors in hypercholesterolemic men and women. J Nutr 134: 2991-2997.
  100.  Ramsden CE, Ringel A, Feldstein AE, Taha AY, MacIntosh BA, et al. (2012) Lowering dietary linoleic acid reduces bioactive oxidized linoleic acid metabolites in humans. Prostaglandins Leukot Essent Fatty Acids 87: 135-141.
  101.  Brymora A, FlisiÅ„ski M, Johnson RJ, Goszka G, StefaÅ„ska A, et al. (2012) Low-fructose diet lowers blood pressure and inflammation in patients with chronic kidney disease. Nephrol Dial Transplant 27: 608-612. [Crossref]
  102.  Gasbarrini G, Mangiola F (2014) Wheat-related disorders: A broad spectrum of 'evolving' diseases. United European Gastroenterol J 2: 254-262. [Crossref]

Article Type

Review Article

Publication history

Received date: March 15, 2017
Accepted date: April 25, 2017
Published date: April 27, 2017

Copyright

© Casas R.This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Casas R (2017) Circulating immune cell activation and diet: A review on human trials. J Allergy Immunol, 2017 DOI: 10.15761/JAI.1000102

Corresponding author

Dr. Ramon Estruch

Department of Internal Medicine, Hospital Clinic, Villarroel, 170, 08036 Barcelona, Spain, Tel. + 34-93-2275745

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

No Figures