Diets link metabolic syndrome and colorectal cancer development (Review)

  • Authors:
    • Jirakrit Saetang
    • Surasak Sangkhathat
  • View Affiliations

  • Published online on: January 18, 2017     https://doi.org/10.3892/or.2017.5385
  • Pages: 1312-1320
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Diets have been believed to be an important factor in the development of metabolic syndrome and colorectal cancer (CRC). In recent years, many studies have shown an intimate relationship between mucosal immunity, metabolism and diets, which has led to a greater understanding of the pathophysiology of metabolic syndrome and CRC development. Although the precise effects of diets on oncogenesis have not been compl­etely elucidated, microbiota changes and inflammation are believed to be important factors that influence the development of CRC. Moreover, increased release of pro-inflammatory cytokines and alteration of adipokine levels have been observed in patients with colorectal adenoma and/or CRC, and these all have been considered as the important mechanisms that link diets to the development of metabolic syndrome and CRC. Importantly, a high-fat, low-fiber diet is associated with dysbiosis, and as the gut signature becomes more important in metabolic syndrome and CRC, an increased understanding of diets on bacterial activity in the pathogenesis of metabolic syndrome and CRC will lead to new preventive and therapeutic strategies.

Introduction

Visceral obesity or, in its broader terminology, metabolic syndrome is an emerging lifestyle-related health problem that has globally increased in prevalence in recent decades (15). Metabolic syndrome is clinically defined as a combination of visceral obesity, increased blood pressure, insulin resistance and dyslipidemia (6,7). This condition has been extensively investigated due to its correlation with various health problems in humans, including cardiovascular diseases, type 2 diabetes (T2D) and non-alcoholic fatty liver disease. In addition to these diseases, which are directly related to visceral adiposity, recent studies have also explored other consequences of metabolic syndrome, such as thromboembolic conditions, hypogonadism and various cancers (811). Recent studies have suggested that the pathophysiology of metabolic syndrome is partly associated with dysfunction of the intestinal flora (1214), a condition called dysbiosis (15). Both brain functions and gut microbiota influence fat intake and increased body mass, which in turn is a source of cytokines that induce chronic, low-grade inflammation and carcinogenesis (1618). Additionally, obesity itself influences the ecology of the gut microbiota population (19).

Colorectal cancer (CRC) is one the cancers that has been associated with metabolic syndrome. CRC is an epithelial cell-derived adenocarcinoma that develops through a multistep carcinogenic process known as the adenoma-carcinoma sequence. Epidemiological studies have demonstrated that colorectal adenomas and CRC risk are higher in individuals with metabolic syndrome or any of its components (2022). A recent meta-analysis of cohort studies reported that metabolic syndrome was associated with higher relative risks of CRC in men at 1.25 (95% confidence interval; 1.19–1.32) and in women at 1.34 (1.09–1.64) (22). In recent decades, partly because of the improved resolution of high-throughput sequencing technology, the gut microbiome has been extensively investigated. Studies have suggested a link between diet-induced microbiota dysbiosis and colorectal carcinogenesis (22).

This review aims to update the current understanding of metabolic syndrome and its role as a pathogenic factor in colorectal pathogenesis.

Metabolic syndrome is a brain-, inflammatory- and microbial-related disease

Metabolic syndrome as a brain disease

Obesity is a major risk factor for several types of cardiovascular and metabolic diseases, such as hyperglycemia, T2D, hypertension and atherosclerosis (23). Individuals with more than three of these symptoms with or without obesity are diagnosed with ‘metabolic syndrome’. Although obesity and metabolic syndrome are generally considered a problem of the gut, neurobiological studies have shown that the brain also plays an important role since neurobiological diseases, such as stroke, dementia, intracranial hypertension and sleep disorders are associated with obesity (24). Moreover, obesity can be initiated by pathological conditions of the brain and several neuropsychiatric drugs (25,26). Therefore, the brain appears to be a central regulator that directs energy fluxes within an organism.

According to the ‘selfish brain theory’ (27) which states that the human brain fulfills its own comparably high energy needs with the highest priority, there are three types of behavioral components regulated by the central nervous system for energy requests: allocative, ingestive and exploratory behaviors (28). The allocation of energy from the body to the central nervous system is the ‘allocative behavior’ carried out by organs such as the liver, pancreas, muscles and adipose tissue (28). The second strategy is ‘ingestive behavior’, describing a situation where the brain acquires energy from its immediate environment, which is initiated by sensory metabolic input signals, including leptin and insulin (2830), and afferent nerve fibers (31,32). The last component, exploratory behavior, enables foraging in distant locations when energy resources in the immediate vicinity become scarce. The brain integrates all three of these behaviors to introduce a feedback control for an ‘energy on demand’ process that maintains an adequate ATP concentration in the nerve cells of the brain. However, if this allocative behavior of the brain fails to satisfy its energy requirements, ingestive behavior increases, leading to the promotion of higher body mass.

Obesity development may be caused by this allocative defect, in which the brain requires energy from food consumption instead of blood sugar (28). The mechanism of this failure may result from an abnormality in various regions of the brain, such as the hypothalamus, amygdala or hippocampus. Clinical studies dating back more than a century have demonstrated that hyperphagia and obesity can be induced by lesions of the ventromedial hypothalamus (VMH), a part of the brain that plays a role in preserving body mass (28,33). In animal studies, lesions of the VMH have caused weight gain in dogs, while VMH stimulation led to satiety (28,34). It is possible that lesions of the VMH lead to increased parasympathetic activity, resulting in an inadequate allocation of glucose to the brain. Then, the animal becomes hyperinsulinemic, finally resulting in hypoglycemia, neuroglycopenia and excessive body mass (3537). In addition, several defective signaling pathways in the hypothalamus have been reported to be involved in obesity development. For example, studies have shown that loss of the leptin receptors in the hypothalamus resulted in obesity, insulin resistance and diabetes in mice (38,39). Leptin signaling abnormalities have also been found in obese patients and were caused by a monogenetic defect or a leptin receptor mutation in the hypothalamus (40,41).

A regulator of insulin and leptin signaling, protein tyrosine phosphatase 1B, has also been associated with leptin and insulin resistance in hypothalamic neurons (42,43) as neuronal protein tyrosine phosphatase 1B-knockout mice showed improved glucose tolerance following chronic high-fat feeding (44). Moreover, VMH lesions may be associated with an unusual vagus nerve-hypothalamus interaction pathway leading to miscommunication between the adipose tissue and β-cells (45). Other factors that result in obesity include a lack of α-melanocyte-stimulating hormone (46), insulin (47) or interleukin-6 (IL-6) (48) or bilateral lesions of the amygdala (36,49). However, not all obese individuals develop obesity due to lesions of the brain. The core concept of the selfish brain theory, the brain-pull hypothesis, posits that ‘brain-pull’ is the force with which the brain actively demands energy from the body (50), indicating that the brain has an active approach associated with specific brain-pull mechanisms (51). Stress has been suggested as the major mechanism that causes the brain to enter a hypervigilant state. This leads to an increased requirement for cerebral energy and initiation of brain-pull mechanisms (52,53), such as ‘cerebral insulin suppression’ that over time results in the loss of subcutaneous adipose tissue, the accumulation of visceral fat and an increased cerebral glucose supply (51).

Metabolic syndrome as an inflammatory disease

Metabolic syndrome and obesity are related to a chronic, low-grade systemic inflammatory condition characterized by increasing levels of circulating inflammatory cytokines and free fatty acids (FFAs) (54). In contrast to classical inflammation, this ‘metabolic inflammation’ (55,56) does not necessarily involve pathogens but occurs as a result of metabolic homeostatic abnormalities. Nowadays, obesity has been associated with inflammatory adipose tissue, which may be caused by higher immune cell accumulation in the fatty tissue region (57). The immune cells associated with this metabolic inflammation include macrophage, neutrophil and lymphocyte.

Adipose tissue macrophages (ATMs)

ATMs appear to play a major role in the regulation of obesity-induced inflammation as ATMs generally play a key role in most cases of chronic inflammation, and these cells can comprise up to 40% of the cells in the adipose tissue (58). It is possible that obesity-induced inflammation is regulated by macrophages (59), and ATM accumulation can be found after as little as 1 week on a high-fat diet (60). Moreover, numerous studies have indicated that accumulation of monocytes and macrophages is positively linked to subcutaneous fat accumulation and weight gain (61). Elevated monocyte levels can be found in the circulation of obese individuals, and the levels decrease when the fat is lost (62,63). These circulating monocytes can be attracted to recruit into the adipose tissue by macrophage chemoattractant protein-1 (MCP-1) (also as known as CCL2), which is secreted by enlarging adipocytes (64,65). Interestingly, mice that lack CCR2, the receptor for MCP-1, showed a decreased food intake and obesity development and increased eosinophil accumulation, which highly increase the expression of IL-4 and IL-13 in the adipose tissue during high-fat diet feeding, leading to the predominance of M2 macrophages (66,67). In addition to CCL2, other studies have found that CXCL12, CXCL14 and CCL5 also play roles in macrophage migration (6870).

In the tissues, macrophages can be subgrouped into 2 types: the classically activated macrophages, termed M1 and the alternatively activated macrophages, termed M2 (71). The M1 macrophage is a CD11c+ pro-inflammatory cell that produces inducible nitric oxide synthase and inflammatory cytokines, such as IL-6 and tumor necrosis factor-α (TNF-α), while the M2 macrophages, which can be identified by the markers CD206, CD209 and CD301 (Mgl1/2), express arginase 1 (Arg1) and immunosuppressive cytokines, including IL-10, transforming growth factor-β (TGF-β), IL-1 receptor antagonist (IL-1RA)-α and IL-4 (59). Interestingly, in obese adipose tissue, the production of anti-inflammatory IL-10 is decreased, and increased levels of inflammatory TNF-α are observed (72). This may be the obvious evidence showing the dominance of the pro-inflammatory macrophage in adipose tissue of obese person. However, recent study demonstrated that inflammatory ATMs found in obese adipose tissue have a uniquely distinct phenotype from M1 macrophages called ‘metabolically activated (MMe) macrophages’ (73), and this phenotype can be induced in vitro by treating macrophages with palmitate, insulin and glucose (metabolic activation) (73). While M1 macrophages display cellular markers CD319, CD274 and CD38, ATMs from obese individuals express lipid transport proteins ATP-binding cassette member 1 (ABCA1) and fatty acid translocase (CD36) (73). Although, both types of macrophages have different phynotypes, they still shared the pro-inflammatory properties by producing TNF-α, IL-6 and IL-1β (73).

Neutrophil

Neutrophils are granulocytes that carry a large number of granules (intracellular vesicles), which contain anti-microbial agents, including lysozyme, neutrophil elastase (NE), myeloperoxidase (MPO) and defensins (59). Although neutrophils are known to play a role in inflammation, it is still unclear whether this type of immune cells is involved in obesity-induced inflammation. Several studies have found that neutrophil migration into the adipose tissue and classical inflammation occurred within 3 days of beginning a high-fat diet in mice (74,75). However, another study found that the neutrophils decreased after 1 week of high-fat feeding, which suggests that neutrophils may play different roles in different stages of obesity development. In this study, they appeared to induce early polarization of ATMs toward the M1 phenotype with corresponding increases in TNF-α and IL-6 (74). Increased percentages of peripheral blood neutrophils have also been found in obese males (76).

T lymphocyte

Lymphocytes are strongly associated with inflammatory processes in obesity. Although there are several lymphocyte cell types that are related to obesity and metabolic syndrome, pro-inflammatory Th1, Th17 and CD8+ T cells predominate over anti-inflammatory regulatory T cells (Treg) and Th2 cells, which are found in higher proportions in lean adipose tissue (77,78). One study found that mice fed a high-fat diet displayed more Th1 polarization and interferon-γ (IFN-γ)production, which occurred several months after macrophage accumulation and insulin resistance (79). However, the number of Treg cells was decreased in the adipose tissue of obese mice, but insulin sensitivity can also be improved when these cells are increased (78). An increased number of Th17 cells in the spleen has been found in mice fed a high-fat diet, but this cell was absent in IL-6−/− mice fed the same high-fat diet, indicating that IL-6 is important for Th17 expansion in obesity (80).

CD8+ T cells are another type of lymphocytes that may be involved in obesity development since co-cultures of splenic CD8+ T cells with adipose tissue from high-fat diet mice showed a higher proliferation rate of CD8+ T cells than those of the lean controls (81). Several studies have also found that higher numbers of CD8+ T cells were positively correlated with an increased risk of insulin resistance (78,81,82). The recovery of CD8+ T cells in knockout mice exacerbated obesity-induced insulin resistance, and this was correlated with increased M1 macrophages and IL-6 and TNF-α gene expression in adipose tissue (81).

B lymphocyte

In addition to T cells, B cells have also shown a positive association with obesity in high-fat diet-fed mice because infiltration of B cells can be found in the adipose tissue of obese mice, and these cells can activate T cells and polarize macrophages toward the M1 type (83). One study found that the number of IgG-associated B cells increased rapidly after 4 weeks on a high-fat diet (84), and the cell numbers then remained consistent during the weight gain (83,85). By using neutralizing antibodies against the B cells, other studies have found that obesity-induced insulin resistance in mice could be improved, and a reduction in M1 macrophages and activation of CD8+ T cells along with an increase in Treg numbers and M2 signatures could be observed in the adipose tissue (83,86). Although B cells are associated with the inflammatory condition in obesity, regulatory B cells, which produce IL-10, have been shown to improve insulin resistance and reduce pro-inflammatory cytokine production in the skeletal muscles of mice fed a high-fat diet (83). These results support the findings from other studies, which found that low levels of IL-10 were associated with metabolic syndrome and T2D (87,88).

Metabolic syndrome as a microbial-related disease

The human intestine contains a unique group of microorganisms comprising up to 1,000 different species with ~100 trillion cells of bacteria, yeasts and parasites (89). There are over 50 bacterial phyla that comprise the gastrointestinal (GI) microbiota, but ~90% consists of Bacteroidetes and Firmicutes (90,91), and diet is clearly seemed to be a major influence on microbiota compositions.

Various evidences have shown that alteration of the diet can result in changes in the bacterial composition that also affected gut metabolic activity, especially the production of short-chain fatty acids (Fig. 1) (92). This scenario can be found in obese individuals (93) as high-fat diets or diets low in fiber have been associated with a higher abundance of Firmicutes (94). Studies comparing obese individuals and their lean twins have also shown a higher predominance of Firmicutes and lower abundance of Bacteroidetes (93,95) in the obese subjects. Other studies, however, have not found a difference (96,97).

Alternatively, the association studies between gut microbiota in obesity and metabolic syndrome have been performed by using germ-free mice. Researchers found that conventionalization of germ-free C57BL/6 mice with normal microbiota harvested from conventionally raised, genetically obese mice resulted in 60% higher levels of body fat and the development of insulin resistance in 2 weeks, despite reduced food consumption (98). Furthermore, a large human study found that low bacterial richness showed a positive correlation with higher weight gain supporting the concept that some bacteria may be more abundant than other types in obese individuals (99). Another study also found that obesity could be transmissible because microbiota derived from discordant obese twins caused germ-free mice to become obese when they were transplanted with these microbiota (100). This study also demonstrated that diet played a major role in gut microbiota diversity because the gut of low-fat, high-fiber diet-fed obese mice was finally dominated by the lean microbiota, which prevented increased adiposity, even when the mice were cohoused with mice with obese microbiota, suggesting that diet is responsible for the development of this phenotype (100).

Colonic carcinogenesis may be promoted by diet-related carcinogenic metabolites

CRC is a common, worldwide cancer causing up to 690,000 deaths/year (101). Although the exact etiology of CRC remains unknown, gradual accumulation of genetic mutations are essential for CRC tumorigenesis (102).

It is clear that the incidence of CRC is higher in developed countries, but in recent years, it has also become a public health problem in developing countries due to economic growth and an increasingly westernized lifestyle, including high consumption of red meats and high-calorie diets, which are associated with an increased incidence of CRC (103,104). This dietary pattern of higher consumption of animal protein (Western diet) that is related to the epidemiology of CRC demonstrates the role of diet in CRC development.

The composition and activity of the intestinal microbiota can be influenced by diet (105), and there is considerable evidence linking diet-induced microbiota dysbiosis to CRC tumorigenesis (106) (Fig. 2). This relationship may be mediated by metabolites and inflammatory induction, which induce genetic mutations and inhibit apoptosis or stimulate angiogenesis and cell proliferation (107). One study found that the increased consumption of red meat was associated with higher levels of sulfur, nitrates, ammonia, amines, branched-chain amino acids, and H2S in the colon (108), which all have been shown to be pro-inflammatory and genotoxic, that in turn facilitates colon tumorigenesis by causing DNA damage and inhibiting the oxidation of butyrate (109,110). Moreover, several species of Bacteroides and some Firmicutes have shown the ability to ferment aromatic amino acids, leading to the production of toxic compounds, such as phenylacetic acid, phenols, indoles, and p-cresol, that promote colon carcinogenesis by DNA alkylation and mutations (111).

Lipid metabolism byproducts, such as bile acids, appear to be associated with the development of CRCs. Secondary bile acids, such as deoxycholic acid (DCA), are involved in the higher levels of reactive oxygen species (ROS), DNA damage, genomic instability and tumor growth (112,113). Moreover, an increased risk of CRC via the higher secondary bile acid production and ROS has been found in individuals consuming a high-fat, low-fiber diet who have a higher composition of inflammatory 7α-dehydroxylating bacteria and sulfur-reducing bacteria, which produce H2S and secondary bile acids, respectively (109). Nitric oxide is another compound produced by specific types of bacteria. Its secondary reactive species can activate inflammation and damage DNA. An investigation by Sobko et al demonstrated that nitrite and nitrate-supplemented diets are sources for nitric oxide production by Lactobacilli and Bifidobacteria, leading to colonic mucosal inflammation and DNA damage (114,115).

The barrier dysfunction mediated by microbial products during CRC development also leads to adenoma invasion by stimulating inflammatory cytokines, such as IL-17 and IL-23, which, in turn, support tumor growth (116). The colonic inflammation model has also demonstrated the loss of the mucosal barrier in dextran sulfate sodium (DSS)-induced colitis. However, when feeding rats with cheese whey protein, colitis markers were reduced, and fecal mucin, a family of glycosylated proteins that play a role in the intestinal mucus layer, and fecal Lactobacilli and Bifidobacteria counts were then increased (117).

Adipocytokines link metabolic syndrome to the risk of CRC

Numerous growth factors, hormones and cytokines, known as adipocytokines, can be secreted by adipose tissue and have pleiotropic effects, including regulation of food intake and metabolism, crosstalk with insulin signaling and inflammatory pathways, and promotion of angiogenesis and cellular proliferation (118,119). These adipocytokines include leptin, resistin, adiponectin and various cytokines, such as TNF-α and IL-6. In obese patients, the levels of these adipocytokines are altered, further contributing to an increased risk of CRC (120,121) (Fig. 2).

Adiponectin

Adiponectin, also known as Acrp30, ADIPOQ, apM1 or GBP28, is a 30 kDa protein that shares homologies with complement factors and TNF-α, and is predominantly secreted by adipocytes (122124). This protein stimulates insulin secretion, increases fatty acid metabolism, activates energy consumption, and is also believed to have an anti-inflammatory role (124,125). The results from epidemiological and preclinical studies have indicated a protective role of adiponectin against tumorigenesis because the levels of adiponectin were negatively correlated with the risk of CRC development (126,127). Recent meta-analyses have shown that, in men, a 1 µg/ml increase in adiponectin levels was associated with a 2% reduced risk of CRC (128130). Studies in animal models have also shown that adiponectin is involved in the suppression of colon carcinogenesis. High-fat diet-fed mice had significantly more polyps when the adiponectin gene was suppressed (131). Moreover, in vitro experiment showed that the growth of CRC cells can be inhibited by adiponectin through the activation of adenosine monophosphate-activated protein kinase (AMPK) and suppression of mammalian target of rapamycin (mTOR) pathways (132). As anti-angiogenic function, neovascularization suppression can be found in adiponectin-treated mice fed a high-fat diet, resulting in significant inhibition of CRC cell growth (133). Adiponectin also showed immunoregulatory functions because this protein induces macrophage infiltration in tumor area (134).

Leptin

Leptin is an adipocyte-derived Ob gene product that plays a major role in the modulation of appetite and energy homeostasis. This is through the feedback regulation between the central nervous system and the peripheral adipose tissue (135,136). Leptin levels appear to be positively associated with obesity and CRC (137,138), and they are believed to be an important factor in the intercellular interactions between adipocytes, fibroblasts, epithelial cells and immune cells (139). An investigation of 108 CRC patients indicated a relationship between leptin levels and progression of cancer, such as lymph node metastasis, distant metastasis and differentiation, suggesting that leptin is involved in cancer proliferation and apoptosis, which may be through the PI3K/Akt/mTOR signaling pathway (140). Leptin also has an anti-apoptotic role in human colon cancer cell lines and promotes the proliferation of normal colonic epithelial and cancer cells as the Wnt signaling pathway, a key pathway in CRC, can be activated by leptin, leading to CRC cell proliferation in vitro (141), while leptin-deficient mice showed a dramatic inhibition of tumor growth (142). Moreover, leptin can also promote CRC progression by stimulating VEGF secretion through hypoxia-inducible factor-1 (HIF-1) and nuclear factor-κB (NF-κB) activation (17,143). Interestingly, it is clear that leptin and adiponectin can neutralize each other's proliferative function because leptin plays a role in blocking the anti-proliferative effects of adiponectin in cancer cells (144), while adiponectin has a negative effect on the proliferative function of leptin (145). This suggests that the cancer risk related to metabolic syndrome can be accurately assessed by the leptin-to-adiponectin ratio.

TNF-α

TNF-α levels in systemic and adipose tissue are higher in obese individuals than in lean subjects (145). Although this cytokine has a functional role in apoptosis, cell necrosis, tumorigenesis inhibition, and appetite reduction (143), its pro-inflammatory role now has been associated with all steps of tumorigenesis (145). High levels of TNF-α in the colonic mucosa of high-fat diet mice resulted in β-catenin activation, leading to increased transcription of c-Myc, a downstream molecule in the Wnt signaling pathway (146). Moreover, this pro-inflammatory cytokine can activate NF-κB, a transcription factor associated with cell proliferation, apoptosis inhibition, tissue invasion, angiogenesis and metastasis, implying that this stimulation is involved in the progression of carcinogenesis (147,148).

Insulin and insulin-like growth factors (IGFs)

Although insulin and IGFs are produced by β-cells in the pancreas and liver, respectively, not in adipose tissue, these factors still play a role in CRC cell proliferation and the progression of cancer both in vitro and in vivo. Circulating total IGF-1 has been positively associated with obesity and thus also contributes to an increased CRC risk (149); one study showed that IGF receptors were found in CRC cells (150). Importantly, several investigations have demonstrated that by binding to these receptors, insulin stimulates proliferation of cultured colonocytes and CRC cells directly, leading to the activation of the MAPK pathway (151,152). This scenario was supported in a study that used monoclonal antibodies against IGF1 and found inhibition of CRC stem cells in mice (153).

Conclusion

Over the last centuries, diets have been shown to affect the pathophysiological development of metabolic syndrome and CRC. Although the precise mechanisms have not been clearly elucidated, it is believed that intestinal inflammation and alteration of adipokines, which may result from a high-fat, low-fiber diet, plays a key role in these processes. This type of diet influences the metabolic activity of the gut and changes the composition of the microbiota, resulting in inflammation and carcinogenesis. Importantly, further investigations are needed for a deeper understanding of the effect of these abnormalities on metabolic imbalance and CRC development. This may lead to the development of new strategies that improve or even treat metabolic syndrome and CRC.

References

1 

Balkau B, Deanfield JE, Després JP, Bassand JP, Fox KA, Smith SC Jr, Barter P, Tan CE, Van Gaal L, Wittchen HU, et al: International Day for the Evaluation of Abdominal Obesity (IDEA): a study of waist circumference, cardiovascular disease, and diabetes mellitus in 168,000 primary care patients in 63 countries. Circulation. 116:1942–1951. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Grundy SM: Metabolic syndrome pandemic. Arterioscler Thromb Vasc Biol. 28:629–636. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Prasad DS, Kabir Z, Dash AK and Das BC: Prevalence and risk factors for metabolic syndrome in Asian Indians: a community study from urban Eastern India. J Cardiovasc Dis Res. 3:204–211. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Gu D, Reynolds K, Wu X, Chen J, Duan X, Reynolds RF, Whelton PK and He J: InterASIA Collaborative Group: Prevalence of the metabolic syndrome and overweight among adults in China. Lancet. 365:1398–1405. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Reynolds K and He J: Epidemiology of the metabolic syndrome. Am J Med Sci. 330:273–279. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Wilson PW: Metabolic risk factors for coronary heart disease: current and future prospects. Curr Opin Cardiol. 14:176–185. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Miranda PJ, DeFronzo RA, Califf RM and Guyton JR: Metabolic syndrome: definition, pathophysiology, and mechanisms. Am Heart J. 149:33–45. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Dentali F, di Minno MN, Gianni M, di Minno G, Squizzato A and Ageno W: The role of the metabolic syndrome in patients with provoked venous thromboembolic events. Thromb Haemost. 109:759–761. 2013. View Article : Google Scholar : PubMed/NCBI

9 

van Rooy MJ, Duim W, Ehlers R, Buys AV and Pretorius E: Platelet hyperactivity and fibrin clot structure in transient ischemic attack individuals in the presence of metabolic syndrome: a microscopy and thromboelastography study. Cardiovasc Diabetol. 14:862015. View Article : Google Scholar : PubMed/NCBI

10 

Ebrahimi F and Christ-Crain M: Metabolic syndrome and hypogonadism - two peas in a pod. Swiss Med Wkly. 146:w142832016.PubMed/NCBI

11 

Uzunlulu M, Telci Caklili O and Oguz A: Association between metabolic syndrome and cancer. Ann Nutr Metab. 68:173–179. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Chen J, He X and Huang J: Diet effects in gut microbiome and obesity. J Food Sci. 79:R442–R451. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Festi D, Schiumerini R, Eusebi LH, Marasco G, Taddia M and Colecchia A: Gut microbiota and metabolic syndrome. World J Gastroenterol. 20:16079–16094. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Parekh PJ, Balart LA and Johnson DA: The Influence of the gut microbiome on obesity, metabolic syndrome and gastrointestinal disease. Clin Transl Gastroenterol. 6:e912015. View Article : Google Scholar : PubMed/NCBI

15 

Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, Quraishi MN, Kinross J, Smidt H, Tuohy KM, et al: The gut microbiota and host health: a new clinical frontier. Gut. 65:330–339. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Pigrau M, Rodiño-Janeiro BK, Casado-Bedmar M, Lobo B, Vicario M, Santos J and Alonso-Cotoner C: The joint power of sex and stress to modulate brain-gut-microbiota axis and intestinal barrier homeostasis: implications for irritable bowel syndrome. Neurogastroenterol Motil. 28:463–486. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Aballay LR, Eynard AR, Díaz M, del P, Navarro A and Muñoz SE: Overweight and obesity: a review of their relationship to metabolic syndrome, cardiovascular disease, and cancer in South America. Nutr Rev. 71:168–179. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Welty FK, Alfaddagh A and Elajami TK: Targeting inflammation in metabolic syndrome. Transl Res. 167:257–280. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD and Gordon JI: Obesity alters gut microbial ecology. Proc Natl Acad Sci USA. 102:11070–11075. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Pischon T, Lahmann PH, Boeing H, Friedenreich C, Norat T, Tjønneland A, Halkjaer J, Overvad K, Clavel-Chapelon F, Boutron-Ruault MC, et al: Body size and risk of colon and rectal cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC). J Natl Cancer Inst. 98:920–931. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Pelucchi C, Negri E, Talamini R, Levi F, Giacosa A, Crispo A, Bidoli E, Montella M, Franceschi S and La Vecchia C: Metabolic syndrome is associated with colorectal cancer in men. Eur J Cancer. 46:1866–1872. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Esposito K, Chiodini P, Colao A, Lenzi A and Giugliano D: Metabolic syndrome and risk of cancer: a systematic review and meta-analysis. Diabetes Care. 35:2402–2411. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Kyrou I, Randeva HS and Weickert MO: Clinical problems caused by obesity: Endotext (Internet). De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, et al: MDText.com, Inc.; South Dartmouth, MA, USA: 2000 https://www.endotext.orgApril 24–2014

24 

Knecht S, Ellger T and Levine JA: Obesity in neurobiology. Prog Neurobiol. 84:85–103. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Sheth RD: Metabolic concerns associated with antiepileptic medications. Neurology. 63 Suppl 4:S24–S29. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Broberger C: Brain regulation of food intake and appetite: molecules and networks. J Intern Med. 258:301–327. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Peters A, Schweiger U, Pellerin L, Hubold C, Oltmanns KM, Conrad M, Schultes B, Born J and Fehm HL: The selfish brain: competition for energy resources. Neurosci Biobehav Rev. 28:143–180. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Peters A, Pellerin L, Dallman MF, Oltmanns KM, Schweiger U, Born J and Fehm HL: Causes of obesity: looking beyond the hypothalamus. Prog Neurobiol. 81:61–88. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Woods SC, Stein LJ, McKay LD and D Jr Porte: Suppression of food intake by intravenous nutrients and insulin in the baboon. Am J Physiol. 247:R393–R401. 1984.PubMed/NCBI

30 

Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E and Flier JS: Role of leptin in the neuroendocrine response to fasting. Nature. 382:250–252. 1996. View Article : Google Scholar : PubMed/NCBI

31 

Kreier F, Kap YS, Mettenleiter TC, van Heijningen C, van der Vliet J, Kalsbeek A, Sauerwein HP, Fliers E, Romijn JA and Buijs RM: Tracing from fat tissue, liver, and pancreas: a neuroanatomical framework for the role of the brain in type 2 diabetes. Endocrinology. 147:1140–1147. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Uno K, Katagiri H, Yamada T, Ishigaki Y, Ogihara T, Imai J, Hasegawa Y, Gao J, Kaneko K, Iwasaki H, et al: Neuronal pathway from the liver modulates energy expenditure and systemic insulin sensitivity. Science. 312:1656–1659. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Dhillon H, Zigman JM, Ye C, Lee CE, McGovern RA, Tang V, Kenny CD, Christiansen LM, White RD, Edelstein EA, et al: Leptin directly activates SF1 neurons in the VMH, and this action by leptin is required for normal body-weight homeostasis. Neuron. 49:191–203. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Brown FD, Fessler RG, Rachlin JR and Mullan S: Changes in food intake with electrical stimulation of the ventromedial hypothalamus in dogs. J Neurosurg. 60:1253–1257. 1984. View Article : Google Scholar : PubMed/NCBI

35 

Choi S, Sparks R, Clay M and Dallman MF: Rats with hypothalamic obesity are insensitive to central leptin injections. Endocrinology. 140:4426–4433. 1999. View Article : Google Scholar

36 

Grundmann SJ, Pankey EA, Cook MM, Wood AL, Rollins BL and King BM: Combination unilateral amygdaloid and ventromedial hypothalamic lesions: evidence for a feeding pathway. Am J Physiol Regul Integr Comp Physiol. 288:R702–R707. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Majdic G, Young M, Gomez-Sanchez E, Anderson P, Szczepaniak LS, Dobbins RL, McGarry JD and Parker KL: Knockout mice lacking steroidogenic factor 1 are a novel genetic model of hypothalamic obesity. Endocrinology. 143:607–614. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Bingham NC, Anderson KK, Reuter AL, Stallings NR and Parker KL: Selective loss of leptin receptors in the ventromedial hypothalamic nucleus results in increased adiposity and a metabolic syndrome. Endocrinology. 149:2138–2148. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Coleman DL: Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice. Diabetologia. 14:141–148. 1978. View Article : Google Scholar : PubMed/NCBI

40 

Clément K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, et al: A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature. 392:398–401. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, et al: Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 387:903–908. 1997. View Article : Google Scholar : PubMed/NCBI

42 

Kaszubska W, Falls HD, Schaefer VG, Haasch D, Frost L, Hessler P, Kroeger PE, White DW, Jirousek MR and Trevillyan JM: Protein tyrosine phosphatase 1B negatively regulates leptin signaling in a hypothalamic cell line. Mol Cell Endocrinol. 195:109–118. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Egawa K, Maegawa H, Shimizu S, Morino K, Nishio Y, Bryer-Ash M, Cheung AT, Kolls JK, Kikkawa R and Kashiwagi A: Protein-tyrosine phosphatase-1B negatively regulates insulin signaling in l6 myocytes and Fao hepatoma cells. J Biol Chem. 276:10207–10211. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Bence KK, Delibegovic M, Xue B, Gorgun CZ, Hotamisligil GS, Neel BG and Kahn BB: Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat Med. 12:917–924. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Cox JE and Powley TL: Prior vagotomy blocks VMH obesity in pair-fed rats. Am J Physiol. 240:E573–E583. 1981.PubMed/NCBI

46 

Brüning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, Klein R, Krone W, Müller-Wieland D and Kahn CR: Role of brain insulin receptor in control of body weight and reproduction. Science. 289:2122–2125. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Volkow ND and Wise RA: How can drug addiction help us understand obesity? Nat Neurosci. 8:555–560. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Wallenius V, Wallenius K, Ahrén B, Rudling M, Carlsten H, Dickson SL, Ohlsson C and Jansson JO: Interleukin-6-deficient mice develop mature-onset obesity. Nat Med. 8:75–79. 2002. View Article : Google Scholar : PubMed/NCBI

49 

King BM: Amygdaloid lesion-induced obesity: relation to sexual behavior, olfaction, and the ventromedial hypothalamus. Am J Physiol Regul Integr Comp Physiol. 291:R1201–R1214. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Peters A: The selfish brain: competition for energy resources. Am J Hum Biol. 23:29–34. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Peters A and McEwen BS: Stress habituation, body shape and cardiovascular mortality. Neurosci Biobehav Rev. 56:139–150. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Hitze B, Hubold C, van Dyken R, Schlichting K, Lehnert H, Entringer S and Peters A: How the selfish brain organizes its supply and demand. Front Neuroenergetics. 2:72010.PubMed/NCBI

53 

Kubera B, Hubold C, Zug S, Wischnath H, Wilhelm I, Hallschmid M, Entringer S, Langemann D and Peters A: The brain's supply and demand in obesity. Front Neuroenergetics. 4:42012. View Article : Google Scholar : PubMed/NCBI

54 

Khodabandehloo H, Gorgani-Firuzjaee S, Panahi G and Meshkani R: Molecular and cellular mechanisms linking inflammation to insulin resistance and β-cell dysfunction. Transl Res. 167:228–256. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Cai D and Liu T: Hypothalamic inflammation: a double-edged sword to nutritional diseases. Ann N Y Acad Sci. 1243:E1–E39. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Cai D and Liu T: Inflammatory cause of metabolic syndrome via brain stress and NF-κB. Aging (Albany, NY). 4:98–115. 2012. View Article : Google Scholar

57 

Gregor MF and Hotamisligil GS: Inflammatory mechanisms in obesity. Annu Rev Immunol. 29:415–445. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL and Ferrante AW Jr: Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 112:1796–1808. 2003. View Article : Google Scholar : PubMed/NCBI

59 

Lee BC and Lee J: Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim Biophys Acta. 1842:446–462. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Lynch L, Nowak M, Varghese B, Clark J, Hogan AE, Toxavidis V, Balk SP, O'Shea D, O'Farrelly C and Exley MA: Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity. 37:574–587. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Yoshimura A, Ohnishi S, Orito C, Kawahara Y, Takasaki H, Takeda H, Sakamoto N and Hashino S: Association of peripheral total and differential leukocyte counts with obesity-related complications in young adults. Obes Facts. 8:1–16. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Krinninger P, Ensenauer R, Ehlers K, Rauh K, Stoll J, Krauss-Etschmann S, Hauner H and Laumen H: Peripheral monocytes of obese women display increased chemokine receptor expression and migration capacity. J Clin Endocrinol Metab. 99:2500–2509. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Poitou C, Dalmas E, Renovato M, Benhamo V, Hajduch F, Abdennour M, Kahn JF, Veyrie N, Rizkalla S, Fridman WH, et al: CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss: relationships with fat mass and subclinical atherosclerosis. Arterioscler Thromb Vasc Biol. 31:2322–2330. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Christiansen T, Richelsen B and Bruun JM: Monocyte chemoattractant protein-1 is produced in isolated adipocytes, associated with adiposity and reduced after weight loss in morbid obese subjects. Int J Obes. 29:146–150. 2005. View Article : Google Scholar

65 

Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, Kitazawa S, Miyachi H, Maeda S, Egashira K, et al: MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 116:1494–1505. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, Charo I, Leibel RL and Ferrante AW Jr: CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. 116:115–124. 2006. View Article : Google Scholar : PubMed/NCBI

67 

Bolus WR, Gutierrez DA, Kennedy AJ, Anderson-Baucum EK and Hasty AH: CCR2 deficiency leads to increased eosinophils, alternative macrophage activation, and type 2 cytokine expression in adipose tissue. J Leukoc Biol. 98:467–477. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Kim D, Kim J, Yoon JH, Ghim J, Yea K, Song P, Park S, Lee A, Hong CP, Jang MS, et al: CXCL12 secreted from adipose tissue recruits macrophages and induces insulin resistance in mice. Diabetologia. 57:1456–1465. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Nara N, Nakayama Y, Okamoto S, Tamura H, Kiyono M, Muraoka M, Tanaka K, Taya C, Shitara H, Ishii R, et al: Disruption of CXC motif chemokine ligand-14 in mice ameliorates obesity-induced insulin resistance. J Biol Chem. 282:30794–30803. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Kitade H, Sawamoto K, Nagashimada M, Inoue H, Yamamoto Y, Sai Y, Takamura T, Yamamoto H, Miyamoto K, Ginsberg HN, et al: CCR5 plays a critical role in obesity-induced adipose tissue inflammation and insulin resistance by regulating both macrophage recruitment and M1/M2 status. Diabetes. 61:1680–1690. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Dalmas E, Clément K and Guerre-Millo M: Defining macrophage phenotype and function in adipose tissue. Trends Immunol. 32:307–314. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Lumeng CN, Bodzin JL and Saltiel AR: Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 117:175–184. 2007. View Article : Google Scholar : PubMed/NCBI

73 

Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, Schoenfelt KQ, Kuzma JN, Larson I, Billing PS, et al: Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab. 20:614–625. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Elgazar-Carmon V, Rudich A, Hadad N and Levy R: Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res. 49:1894–1903. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, Lu M, Li P, Yan Q, Zhu Y, et al: Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med. 18:1407–1412. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Xu X, Su S and Wang X, Barnes V, De Miguel C, Ownby D, Pollock J, Snieder H, Chen W and Wang X: Obesity is associated with more activated neutrophils in African American male youth. Int J Obes (Lond). 39:26–32. 2015.PubMed/NCBI

77 

Sell H, Habich C and Eckel J: Adaptive immunity in obesity and insulin resistance. Nat Rev Endocrinol. 8:709–716. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, Lee J, Goldfine AB, Benoist C, Shoelson S, et al: Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 15:930–939. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Strissel KJ, DeFuria J, Shaul ME, Bennett G, Greenberg AS and Obin MS: T-cell recruitment and Th1 polarization in adipose tissue during diet-induced obesity in C57BL/6 mice. Obesity (Silver Spring). 18:1918–1925. 2010. View Article : Google Scholar : PubMed/NCBI

80 

Winer S, Paltser G, Chan Y, Tsui H, Engleman E, Winer D and Dosch HM: Obesity predisposes to Th17 bias. Eur J Immunol. 39:2629–2635. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, Otsu M, Hara K, Ueki K, Sugiura S, et al: CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 15:914–920. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Wu H, Ghosh S, Perrard XD, Feng L, Garcia GE, Perrard JL, Sweeney JF, Peterson LE, Chan L, Smith CW, et al: T-cell accumulation and regulated on activation, normal T cell expressed and secreted upregulation in adipose tissue in obesity. Circulation. 115:1029–1038. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, Tsui H, Wu P, Davidson MG, Alonso MN, et al: B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 17:610–617. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Sun S, Ji Y, Kersten S and Qi L: Mechanisms of inflammatory responses in obese adipose tissue. Annu Rev Nutr. 32:261–286. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Duffaut C, Galitzky J, Lafontan M and Bouloumié A: Unexpected trafficking of immune cells within the adipose tissue during the onset of obesity. Biochem Biophys Res Commun. 384:482–485. 2009. View Article : Google Scholar : PubMed/NCBI

86 

DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR, Markham D, Strissel KJ, Watkins AA, Zhu M, et al: B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci USA. 110:5133–5138. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Jagannathan M, McDonnell M, Liang Y, Hasturk H, Hetzel J, Rubin D, Kantarci A, Van Dyke TE, Ganley-Leal LM and Nikolajczyk BS: Toll-like receptors regulate B cell cytokine production in patients with diabetes. Diabetologia. 53:1461–1471. 2010. View Article : Google Scholar : PubMed/NCBI

88 

van Exel E, Gussekloo J, De Craen AJM, Frölich M, Bootsma-Van Der Wiel A and Westendorp RGJ: Leiden 85 Plus Study: Low production capacity of interleukin-10 associates with the metabolic syndrome and type 2 diabetes: The Leiden 85-Plus Study. Diabetes. 51:1088–1092. 2002. View Article : Google Scholar : PubMed/NCBI

89 

Gomes AC, Bueno AA, de Souza RGM and Mota JF: Gut microbiota, probiotics and diabetes. Nutr J. 13:602014. View Article : Google Scholar : PubMed/NCBI

90 

Bajzer M and Seeley RJ: Physiology: obesity and gut flora. Nature. 444:1009–1010. 2006. View Article : Google Scholar : PubMed/NCBI

91 

Sekirov I, Russell SL, Antunes LCM and Finlay BB: Gut microbiota in health and disease. Physiol Rev. 90:859–904. 2010. View Article : Google Scholar : PubMed/NCBI

92 

David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, et al: Diet rapidly and reproducibly alters the human gut microbiome. Nature. 505:559–563. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al: A core gut microbiome in obese and lean twins. Nature. 457:480–484. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod LP, Harris NL, et al: Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 20:159–166. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Ley RE, Turnbaugh PJ, Klein S and Gordon JI: Microbial ecology: human gut microbes associated with obesity. Nature. 444:1022–1023. 2006. View Article : Google Scholar : PubMed/NCBI

96 

Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C and Hardt PD: Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring). 18:190–195. 2010. View Article : Google Scholar : PubMed/NCBI

97 

Duncan SH, Lobley GE, Holtrop G, Ince J, Johnstone AM, Louis P and Flint HJ: Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes. 32:1720–1724. 2008. View Article : Google Scholar

98 

Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF and Gordon JI: The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 101:15718–15723. 2004. View Article : Google Scholar : PubMed/NCBI

99 

Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M, Batto JM, Kennedy S, et al: MetaHIT consortium: Richness of human gut microbiome correlates with metabolic markers. Nature. 500:541–546. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, et al: Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 341:12412142013. View Article : Google Scholar : PubMed/NCBI

101 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Sears CL and Garrett WS: Microbes, microbiota, and colon cancer. Cell Host Microbe. 15:317–328. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Center MM, Jemal A and Ward E: International trends in colorectal cancer incidence rates. Cancer Epidemiol Biomarkers Prev. 18:1688–1694. 2009. View Article : Google Scholar : PubMed/NCBI

104 

Center MM, Jemal A, Smith RA and Ward E: Worldwide variations in colorectal cancer. CA Cancer J Clin. 59:366–378. 2009. View Article : Google Scholar : PubMed/NCBI

105 

Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ and Lobley GE: Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol. 73:1073–1078. 2007. View Article : Google Scholar : PubMed/NCBI

106 

Keku TO, Dulal S, Deveaux A, Jovov B and Han X: The gastrointestinal microbiota and colorectal cancer. Am J Physiol Gastrointest Liver Physiol. 308:G351–G363. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Medzhitov R: Origin and physiological roles of inflammation. Nature. 454:428–435. 2008. View Article : Google Scholar : PubMed/NCBI

108 

Nyangale EP, Mottram DS and Gibson GR: Gut microbial activity, implications for health and disease: the potential role of metabolite analysis. J Proteome Res. 11:5573–5585. 2012. View Article : Google Scholar : PubMed/NCBI

109 

Greer JB and O'Keefe SJ: Microbial induction of immunity, inflammation, and cancer. Front Physiol. 1:1682011. View Article : Google Scholar : PubMed/NCBI

110 

Módis K, Coletta C, Erdélyi K, Papapetropoulos A and Szabo C: Intramitochondrial hydrogen sulfide production by 3-mercaptopyruvate sulfurtransferase maintains mitochondrial electron flow and supports cellular bioenergetics. FASEB J. 27:601–611. 2013. View Article : Google Scholar : PubMed/NCBI

111 

Nistal E, Fernández-Fernández N, Vivas S and Olcoz JL: Factors determining colorectal cancer: the role of the intestinal microbiota. Front Oncol. 5:2202015. View Article : Google Scholar : PubMed/NCBI

112 

Dvorak K, Payne CM, Chavarria M, Ramsey L, Dvorakova B, Bernstein H, Holubec H, Sampliner RE, Guy N, Condon A, et al: Bile acids in combination with low pH induce oxidative stress and oxidative DNA damage: relevance to the pathogenesis of Barrett's oesophagus. Gut. 56:763–771. 2007. View Article : Google Scholar : PubMed/NCBI

113 

Gill CI and Rowland IR: Diet and cancer: assessing the risk. Br J Nutr. 88 Suppl 1:S73–S87. 2002. View Article : Google Scholar : PubMed/NCBI

114 

Sobko T, Huang L, Midtvedt T, Norin E, Gustafsson LE, Norman M, Jansson EA and Lundberg JO: Generation of NO by probiotic bacteria in the gastrointestinal tract. Free Radic Biol Med. 41:985–991. 2006. View Article : Google Scholar : PubMed/NCBI

115 

Sobko T, Reinders CI, Jansson E, Norin E, Midtvedt T and Lundberg JO: Gastrointestinal bacteria generate nitric oxide from nitrate and nitrite. Nitric Oxide. 13:272–278. 2005. View Article : Google Scholar : PubMed/NCBI

116 

Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, et al: Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 491:254–258. 2012.PubMed/NCBI

117 

Sprong RC, Schonewille AJ and van der Meer R: Dietary cheese whey protein protects rats against mild dextran sulfate sodium-induced colitis: role of mucin and microbiota. J Dairy Sci. 93:1364–1371. 2010. View Article : Google Scholar : PubMed/NCBI

118 

Hursting SD, Digiovanni J, Dannenberg AJ, Azrad M, Leroith D, Demark-Wahnefried W, Kakarala M, Brodie A and Berger NA: Obesity, energy balance, and cancer: new opportunities for prevention. Cancer Prev Res (Phila). 5:1260–1272. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Tilg H and Moschen AR: Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 6:772–783. 2006. View Article : Google Scholar : PubMed/NCBI

120 

Bardou M, Barkun AN and Martel M: Obesity and colorectal cancer. Gut. 62:933–947. 2013. View Article : Google Scholar : PubMed/NCBI

121 

Barb D, Williams CJ, Neuwirth AK and Mantzoros CS: Adiponectin in relation to malignancies: a review of existing basic research and clinical evidence. Am J Clin Nutr. 86:s858–s866. 2007.PubMed/NCBI

122 

Scherer PE, Williams S, Fogliano M, Baldini G and Lodish HF: A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem. 270:26746–26749. 1995. View Article : Google Scholar : PubMed/NCBI

123 

Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K and Tobe K: Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest. 116:1784–1792. 2006. View Article : Google Scholar : PubMed/NCBI

124 

Nakashima R, Kamei N, Yamane K, Nakanishi S, Nakashima A and Kohno N: Decreased total and high molecular weight adiponectin are independent risk factors for the development of type 2 diabetes in Japanese-Americans. J Clin Endocrinol Metab. 91:3873–3877. 2006. View Article : Google Scholar : PubMed/NCBI

125 

Renehan AG, Zwahlen M and Egger M: Adiposity and cancer risk: new mechanistic insights from epidemiology. Nat Rev Cancer. 15:484–498. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Guffey CR, Fan D, Singh UP and Murphy EA: Linking obesity to colorectal cancer: recent insights into plausible biological mechanisms. Curr Opin Clin Nutr Metab Care. 16:595–600. 2013. View Article : Google Scholar : PubMed/NCBI

127 

Hillenbrand A, Fassler J, Huber N, Xu P, Henne-Bruns D, Templin M, Schrezenmeier H, Wolf AM and Knippschild U: Changed adipocytokine concentrations in colorectal tumor patients and morbidly obese patients compared to healthy controls. BMC Cancer. 12:5452012. View Article : Google Scholar : PubMed/NCBI

128 

Xu XT, Xu Q, Tong JL, Zhu MM, Huang ML, Ran ZH and Xiao SD: Meta-analysis: circulating adiponectin levels and risk of colorectal cancer and adenoma. J Dig Dis. 12:234–244. 2011. View Article : Google Scholar : PubMed/NCBI

129 

An W, Bai Y, Deng SX, Gao J, Ben QW, Cai QC, Zhang HG and Li ZS: Adiponectin levels in patients with colorectal cancer and adenoma: a meta-analysis. Eur J Cancer Prev. 21:126–133. 2012. View Article : Google Scholar : PubMed/NCBI

130 

Gulcelik MA, Colakoglu K, Dincer H, Dogan L, Yenidogan E and Gulcelik NE: Associations between adiponectin and two different cancers: breast and colon. Asian Pac J Cancer Prev. 13:395–398. 2012. View Article : Google Scholar : PubMed/NCBI

131 

Fujisawa T, Endo H, Tomimoto A, Sugiyama M, Takahashi H, Saito S, Inamori M, Nakajima N, Watanabe M, Kubota N, et al: Adiponectin suppresses colorectal carcinogenesis under the high-fat diet condition. Gut. 57:1531–1538. 2008. View Article : Google Scholar : PubMed/NCBI

132 

Sugiyama M, Takahashi H, Hosono K, Endo H, Kato S, Yoneda K, Nozaki Y, Fujita K, Yoneda M, Wada K, et al: Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int J Oncol. 34:339–344. 2009.PubMed/NCBI

133 

Moon HS, Liu X, Nagel JM, Chamberland JP, Diakopoulos KN, Brinkoetter MT, Hatziapostolou M, Wu Y, Robson SC, Iliopoulos D, et al: Salutary effects of adiponectin on colon cancer: in vivo and in vitro studies in mice. Gut. 62:561–570. 2013. View Article : Google Scholar : PubMed/NCBI

134 

Sun Y and Lodish HF: Adiponectin deficiency promotes tumor growth in mice by reducing macrophage infiltration. PLoS One. 5:e119872010. View Article : Google Scholar : PubMed/NCBI

135 

Vansaun MN: Molecular pathways: adiponectin and leptin signaling in cancer. Clin Cancer Res. 19:1926–1932. 2013. View Article : Google Scholar : PubMed/NCBI

136 

Friedman JM and Halaas JL: Leptin and the regulation of body weight in mammals. Nature. 395:763–770. 1998. View Article : Google Scholar : PubMed/NCBI

137 

Ryan AS, Berman DM, Nicklas BJ, Sinha M, Gingerich RL, Meneilly GS, Egan JM and Elahi D: Plasma adiponectin and leptin levels, body composition, and glucose utilization in adult women with wide ranges of age and obesity. Diabetes Care. 26:2383–2388. 2003. View Article : Google Scholar : PubMed/NCBI

138 

Tamakoshi K, Toyoshima H, Wakai K, Kojima M, Suzuki K, Watanabe Y, Hayakawa N, Yatsuya H, Kondo T, Tokudome S, et al: Leptin is associated with an increased female colorectal cancer risk: a nested case-control study in Japan. Oncology. 68:454–461. 2005. View Article : Google Scholar : PubMed/NCBI

139 

Delort L, Rossary A, Farges MC, Vasson MP and Caldefie-Chézet F: Leptin, adipocytes and breast cancer: focus on inflammation and anti-tumor immunity. Life Sci. 140:37–48. 2015. View Article : Google Scholar : PubMed/NCBI

140 

Wang D, Chen J, Chen H, Duan Z, Xu Q, Wei M, Wang L and Zhong M: Leptin regulates proliferation and apoptosis of colorectal carcinoma through PI3K/Akt/mTOR signalling pathway. J Biosci. 37:91–101. 2012. View Article : Google Scholar : PubMed/NCBI

141 

Padidar S, Farquharson AJ, Williams LM, Kearney R, Arthur JR and Drew JE: High-fat diet alters gene expression in the liver and colon: links to increased development of aberrant crypt foci. Dig Dis Sci. 57:1866–1874. 2012. View Article : Google Scholar : PubMed/NCBI

142 

Endo H, Hosono K, Uchiyama T, Sakai E, Sugiyama M, Takahashi H, Nakajima N, Wada K, Takeda K, Nakagama H, et al: Leptin acts as a growth factor for colorectal tumours at stages subsequent to tumour initiation in murine colon carcinogenesis. Gut. 60:1363–1371. 2011. View Article : Google Scholar : PubMed/NCBI

143 

Braun S, Bitton-Worms K and LeRoith D: The link between the metabolic syndrome and cancer. Int J Biol Sci. 7:1003–1015. 2011. View Article : Google Scholar : PubMed/NCBI

144 

Grossmann ME, Mizuno NK, Bonorden MJL, Ray A, Sokolchik I, Narasimhan ML and Cleary MP: Role of the adiponectin leptin ratio in prostate cancer. Oncol Res. 18:269–277. 2009. View Article : Google Scholar : PubMed/NCBI

145 

Sharma D, Wang J, Fu PP, Sharma S, Nagalingam A, Mells J, Handy J, Page AJ, Cohen C, Anania FA, et al: Adiponectin antagonizes the oncogenic actions of leptin in hepatocellular carcinogenesis. Hepatology. 52:1713–1722. 2010. View Article : Google Scholar : PubMed/NCBI

146 

Liu Z, Brooks RS, Ciappio ED, Kim SJ, Crott JW, Bennett G, Greenberg AS and Mason JB: Diet-induced obesity elevates colonic TNF-α in mice and is accompanied by an activation of Wnt signaling: a mechanism for obesity-associated colorectal cancer. J Nutr Biochem. 23:1207–1213. 2012. View Article : Google Scholar : PubMed/NCBI

147 

Dolcet X, Llobet D, Pallares J and Matias-Guiu X: NF-κB in development and progression of human cancer. Virchows Arch. 446:475–482. 2005. View Article : Google Scholar : PubMed/NCBI

148 

Pais R, Silaghi H, Silaghi AC, Rusu ML and Dumitrascu DL: Metabolic syndrome and risk of subsequent colorectal cancer. World J Gastroenterol. 15:5141–5148. 2009. View Article : Google Scholar : PubMed/NCBI

149 

Clayton PE, Banerjee I, Murray PG and Renehan AG: Growth hormone, the insulin-like growth factor axis, insulin and cancer risk. Nat Rev Endocrinol. 7:11–24. 2011. View Article : Google Scholar : PubMed/NCBI

150 

Singh P and Rubin N: Insulinlike growth factors and binding proteins in colon cancer. Gastroenterology. 105:1218–1237. 1993. View Article : Google Scholar : PubMed/NCBI

151 

Giovannucci E: Nutrition, insulin, insulin-like growth factors and cancer. Horm Metab Res. 35:694–704. 2003. View Article : Google Scholar : PubMed/NCBI

152 

Komninou D, Ayonote A, Richie JP Jr and Rigas B: Insulin resistance and its contribution to colon carcinogenesis. Exp Biol Med (Maywood). 228:396–405. 2003.PubMed/NCBI

153 

Hart LS, Dolloff NG, Dicker DT, Koumenis C, Christensen JG, Grimberg A and El-Deiry WS: Human colon cancer stem cells are enriched by insulin-like growth factor-1 and are sensitive to figitumumab. Cell Cycle. 10:2331–2338. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 37 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Saetang J and Saetang J: Diets link metabolic syndrome and colorectal cancer development (Review). Oncol Rep 37: 1312-1320, 2017
APA
Saetang, J., & Saetang, J. (2017). Diets link metabolic syndrome and colorectal cancer development (Review). Oncology Reports, 37, 1312-1320. https://doi.org/10.3892/or.2017.5385
MLA
Saetang, J., Sangkhathat, S."Diets link metabolic syndrome and colorectal cancer development (Review)". Oncology Reports 37.3 (2017): 1312-1320.
Chicago
Saetang, J., Sangkhathat, S."Diets link metabolic syndrome and colorectal cancer development (Review)". Oncology Reports 37, no. 3 (2017): 1312-1320. https://doi.org/10.3892/or.2017.5385