Skip to main content

Hydroxycinnamic acid derivatives: a potential class of natural compounds for the management of lipid metabolism and obesity

Abstract

Hydroxycinnamic acid derivatives are important class of polyphenolic compounds originated from the Mavolanate-Shikimate biosynthesis pathways in plants. Several simple phenolic compounds such as cinnamic acid, p-coumaric acid, ferulic acid, caffeic acid, chlorgenic acid, and rosmarinic acid belong to this class. These phenolic compounds possess potent antioxidant and anti-inflammatory properties. These compounds were also showed potential therapeutic benefit in experimental diabetes and hyperlipidemia. Recent evidences also suggest that they may serve as valuable molecule for the treatment of obesity related health complications. In adipose tissues, hydroxycinnamic acid derivatives inhibit macrophage infiltration and nuclear factor κB (NF-κB) activation in obese animals. Hydroxycinnamic acid derivatives also reduce the expression of the potent proinflammatory adipokines tumor necrosis factor-α (TNFα), monocyte chemoattractant protein-1 (MCP-1), and plasminogen activator inhibitor type-1 (PAI-1), and they increase the secretion of an anti-inflammatory agent adiponectin from adipocytes. Furthermore, hydroxycinnamic acid derivatives also prevent adipocyte differentiation and lower lipid profile in experimental animals. Through these diverse mechanisms hydroxycinnamic acid derivatives reduce obesity and curtail associated adverse health complications.

Background

Metabolic syndrome is a cluster of non-communicable diseases includes central obesity, diabetes, insulin resistance, hypertension and dyslipidemia. Prevalence of obesity and diabetes are increasing day by day among children, young and elderly populations both in developed and developing countries [1–3]. A sedentary nature of jobs and high calorie diet mainly western style diet are the main causes of developing obesity and diabetes, consequently metabolic syndrome [1, 4]. Ever increasing obese and diabetes population are causing serious problems for the management of health sector as well as increasing personal health risks [4]. Recent evidence also suggests that increased body fat mass causes cardiovascular diseases and increases morbidity and mortality in human [5, 6]. Dietary modification, for example decreasing the intake of high fat and high carbohydrate could be a possible way of reducing the risk of fat accumulation in the body. In addition, several dietary approaches such as Mediterranean diet or diet containing high amount of fibres, fruits and vegetables would be valuable for the prevention of hypertension, diabetes, dyslipidemia and obesity [7, 8]. Mediterranean diet or fruits and vegetables possess large amount of phenolic or polyphenolic compounds. It is now widely recognized that, phenolic or polyphenolic compounds are strong antioxidant substances and showed anti-inflammatory properties [9, 10]. Some of them are also effective against diabetes, insulin resistance and dyslipidemia [11–15]. Many of them also prevent hypertension and cardiovascular diseases [16]. All these biological activities are mainly regulated by phenolic acid’s ability to scavenge free radicals generated due to excess nutrition supply to the tissues in obesity, or they may regulate the energy homeostasis and inflammatory pathways. This work will thus review the potential health benefit of hydroxycinnamic acid derivatives in obesity and metabolic syndrome and their possible mechanism of action.

Obesity and energy homeostasis, mechanism of fat metabolism

Obesity can be defined as the accumulation of excess fat due to the increased energy intake and lack of energy expenditure. However, World Health Organization uses Body Mass Index (BMI) as a parameter for defining obesity. According to WHO, BMI >30 is considered as moderately obese and BMI > 35 is considered as severely obese in human [17]. Global obesity in young to adult population is increasing tremendously in recent years [18, 19]. Lack of physical movements, sedentary nature of work and consumption of diet containing high carbohydrate and high fat are responsible for the development of obesity [20]. Thus, increased energy expenditure would be a contributing factor to control and manage obesity and related pathophysiological conditions. Mitochondrial biogenesis are the major pathways in various cell types like, liver, adipose tissue, skeletal muscle etc. to increase ATP production and energy expenditure. Decreased mitochondrial function was observed in obesity and metabolic disorder [21–23]. In obese condition, abundance of fuel supply e.g., fatty acid and glucose overwhelm the mitochondrial electron transport chain and increased the superoxide production [24–27]. Mitochondrial biogeneses are regulated via several transcriptional regulatory factors like AMPK, PPAR- γ and PGC-1α [28, 29]. AMPK regulated PPAR-γ and PGC-1α activation stimulated most of the transcriptional signal to increase fatty acid oxidation and mitochondrial function [30–32].

AMPK

AMP-activated protein kinase (AMPK) is a cellular fuel gauge, maintaining intracellular energy balance in mammalian cells [33]. AMPK signalling pathway is activated by elevation of the AMP/ATP ratio due to the decreased ATP synthesis by mitochondria or by increased energy (ATP) expenditure [33]. Glucose deprivation, hypoxia or ischaemia, or metabolic poisons are few factors which may inhibit glycolysis, tricarboxylic acid cycle or oxidative phosphorylation and disturb energy balance by interfering with ATP synthesis, which may trigger activation of AMPK signalling [34]. AMPK activation is necessary for the transcriptional regulation of energy demand. Mice expressing a dominant-negative form of AMPK failed to increase mitochondrial biogenesis in response to energy deprivation in skeletal muscles [35]. In contrast, lipid oxidation and mitochondrial activity was increased in mice over expressing the phosphorylated AMPK [36, 37]. Several ligands such as thiazolidinediones (for example, rosiglitazone) and biguanides (metformin) both activates AMPK [38]. Thiazolidinediones and biguanides inhibits complex I of the mitochondrial respiratory chain and elevates cellular AMP/ATP ratios [39]. Furthermore, mice fed with AMPK agonists increased oxidative gene expression, enhanced endurance capacity and gave protection against metabolic disease [40, 41]. AMPK can also be activated by metabolic stresses such as muscle contraction or hypoxia, and modulated by hormones and cytokines affecting whole-body energy balance such as leptin, adiponectin, resistin, ghrelin and cannabinoids [33].

PPAR- γ

AMPK activation increased the fatty acid oxidation through activating the PPAR-γ and PGC-1α [42]. Peroxisome proliferator activator protein-γ (PPAR- γ) is highly expressed in adipose tissues [43]. The expression of PPARγ in liver is very low compared to the level present in adipose tissue [43–45]. The actions of PPAR-γ are mediated by two protein isoforms, PPARγ1 and PPARγ2 [46]. PPARγ1 is widely expressed while PPARγ2 is restricted to the adipose tissue only [46]. Fatty acids binding activates PPAR-γ [46]. Activation of PPARγ is necessary for adipocyte differentiation and fatty-acid storage [43, 44]. PPAR-γ deficient mice are devoid of adipose tissue and PPAR-γ +/− mice are characterized by a decreased adipose tissue mass [47, 48]. PPAR-γ is also important for anti-inflammatory pathways, lipid metabolism and regulates genes taking part in the release, transport and storage of fatty acids [49, 50]. Moreover, PPARγ is also responsible for the improvement of insulin resistance and plays an important role in glucose homeostasis. Mice lacking PPARγ in fat, muscle, or liver are predisposed to develop insulin resistance [51–54] while mice with increased PPARγ activities are protected from obesity-associated insulin resistance [55]. PPARγ is a ligand activated protein, thiazolidinediones are considered as the activator of PPARγ [56]. However, thiazolidinediones are adipogenic and responsible for moderate weight gain in patients taking thiazolidinediones [50, 57].

PGC-1α

Peroxisome proliferator activator protein-γ co-activator-1α (PGC-1α) is another regulator of lipid and glucose metabolism. AMPK regulates PGC-1α at both gene and protein level [36]. PGC-1α directly co-activates multiple transcriptional factors such as the PPARs or the thyroid hormone receptor, glucocorticoid receptors and estrogen receptors [29, 58]. PGC-1α also increases mitochondrial biogenesis and respiration rates, as well as the uptake and utilization of substrates for energy production [59]. In brown adipose tissue (BAT), cold induces PGC-1α protein expression that controls adaptive thermogenesis [59]. Furthermore, fasting induces hepatic PGC-1α expression and increases gluconeogenesis, whereas in skeletal and cardiac muscle, exercise increases PGC-1α mediated mitochondrial biogenesis and respiration [60]. Phenolic compound resveratrol increased the PGC-1α activity and increased running time and consumption of oxygen in muscle fibers in mice [61]. Moreover, resveratrol increased insulin sensitivity, reduced insulin-like growth factor-1 (IGF-I) levels, increased AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor-gamma co-activator-1α (PGC-1α) activity, increased mitochondrial number, and improved motor function in middle-aged mice fed a high-calorie diet [62].

Inflammation and obesity

Inflammation is a protective response mechanism for tissue injury. Both acute and chronic inflammatory responses are responsible for the development of diabetes and insulin resistance [63, 64]. Recent research findings suggest that chronic low grade inflammation is developed in obese individuals and triggers adipocyte dysfunction [65]. Moreover, adipose tissues are playing a major role in secreting pro-inflammatory and inflammatory cytokines during obesity [66]. Pathologic growth of adipocyte houses many of inflammatory cytokines like TNF-alpha and IL-6 [67]. Inputs into this inflammatory response further stimulate ER stress, adipose tissue hypoxia, and adipocyte death [68–70]. Macrophage numbers in adipose tissues are also increased with obesity where they mainly scavenge the dead adipocytes [71, 72]. Macrophages are also responsible for the cytokine production in obese adipose tissues [73].

Hydroxycinnamic acid derivatives overview

Hydroxycinnamic acid derivatives (Fig. 1) comprise a large group of simple phenolic acids, found mainly in cereals, fruits and vegetables. A review has been published recently describing the occurrence, biosynthesis, and pharmacokinetics of hydroxycinnamic acid derivatives [74]. Ferulic acid, caffeic acid, p-coumaric acid, chlorgenic acid, sinapic acid, curcumin, and rosmarinic acid belongs to this important phenolic acid group. Hydroxycinnamic acids are abundant in fruits, vegetables and cereals and seeds of fruits [74]. In plant, hydroxycinnamic acid derivatives are synthesized by following the mavolonate and shikimate pathways where phenylalanine and tyrosine are two starter precursor molecules [74, 75]. Following several intermediate enzymatic process, shikimate pathways produced cinnamic acid, p-coumaric acid, caffeic acid, ferulic acid, sinapic acid to various complex lignin molecules [74, 75]. Hydroxycinnamic acid derivatives are also serving as precursor molecules for the stilbenes, chalcons, flavonoids, lignans, and anthocyanins [74].

Fig. 1
figure 1

Structures of hydroxyl cinnamic acid derivatives. Cinnamic acid, p-caumaric acid, ferulic acid, caffeic acid, chlorgenic acid, rosemarinic acid

HCAs are absorbed easily from the stomach and intestine depending on their structure and compared to other complex phenolic compounds [76]. Ferulic acid and p-coumaric acid are also absorbed from intestine, jejunum, ileum and colon of rats [74]. However, chlorgenic acid, ester of caffeic and quinic acid, first hydrolysed and free caffeic acid is absorbed from the intestine [76]. In Caco-2 cell monolayers, caffeic acid demonstrated that monocarboxylic acid transporters (MCTs), a transport system present across the intestinal epithelial cells, may be involved in the absorption process [77, 78]. p-Coumaric acid and ferulic acid also followed the same monocarboxylic acid transporter (MCT) system to cross the intestinal epithelium [77, 78]. However, passive diffusion mechanism is also important and not ignored for the absorption of ferulic acid in the stomach and Caco-2 cells [75, 79]. In addition, involvement of a Na+-dependent, carrier-mediated transport process are also involved in the uptake of cinnamic acid and ferulic acid across the brush border membrane of rat jejunum [80]. Bioavailability of cinnamic acid derivatives are reviewed recently [74, 76]. Various cinnamic acid derivatives can be found in plasma immediately after the oral administration and may show various health benefit in different diseases (Fig. 2).

Fig. 2
figure 2

Health benefit of cinnamic acid derivatives in various diseases

Effect of Hydroxycinnamic acid derivatives on various parameters of Metabolic syndrome

Effect of hydroxycinnamic acid derivatives in Inflammation

Hydroxycinnamic acid derivatives showed anti-inflammatory properties both in vitro and in vivo [81]. Ferulic acid prevented the production of TNF-alpha and decreased Macrophage inflammatory protein-2 (MIP-2) levels in lipopolysaccharide (LPS)-stimulated RAW264.7 cells [82]. The transcription factor nuclear factor kappa B (NF-κB) plays a critical role in stress, immune, and inflammatory responses. Ferulic acid in cereals inhibits NF-kB activation [83]. Salt of ferulic acid, ferulate, exhibited antioxidant action by maintaining redox regulation, suppressing NF-κB activation and modulating the expression of NF-κB-induced, proinflammatory COX-2, iNOS, VCAM-1 and ICAM-1 in aged Sprague–Dawley rats [84]. NF-kB suppression by ferulate is mediated via suppressing the activation of NIK/IKK and MAPKs [84].

p-Coumaric acid prevented the increased cell-mediated immune responses and macrophage phagocytic index in rats [85].p- Coumaric acid also decrease in the expression of inflammatory mediator TNF-α and circulating immune complexes in adjuvant induced arthritic rats [85]. p-Coumaric acid also inhibited the TNF-α-induced changes in levels of monocyte chemoattractant protein-1 (MCP-1), plasminogen activator inhibitor-1 (PAI-1), and intracellular reactive oxygen species (ROS) in 3 T3-L1 adipocytes [86]. Furthermore, p-coumaric acid increased the secretion and concentration of adiponectin, superoxide dismutase (SOD), glutathione (GSH), glutathione peroxidase (GPx), and glutathione S-transferase (GST) in TNF-α-treated 3 T3-L1 adipocytes [86].

Caffeic acid phenetyl ester (CAPE) non-selectively inhibited the activities of baculovirus-expressed hCOX-1 and hCOX-2 enzymes and inhibits prostaglandin synthesis and COX 2 in the rat carrageenan air pouch model of inflammation [87]. Caffeic acid and some of its derivatives such as caffeic acid phenetyl ester (CAPE) and octyl caffeate showed anti-inflammatory activity both in vitro and in vivo [88]. Caffeic acid derivatives suppressed the iNOS expression and prevented the production of NO from RAW macrophage cells. Moreover, butyl, octyl and CAPE derivatives of caffeic acid inhibited carrageenan-induced paw edema and prevented the increase in IL-1β levels in the mouse paw [88]. Butyl, octyl and CAPE derivatives also prevented carrageenan-induced neutrophil influx in the mouse paw [88]. Caffeic acid supplementation reduced the inflammatory cytokines interleukin (IL)-beta, IL-6, tumor necrosis factor (TNF)-alpha and monocyte chemoattractant protein (MCP)-1 concentration in diabetic mice [16].

Chlorogenic acid, the ester of caffeic acid with quinic acid blocked UVB- or TPA-induced transactivation of AP-1 and NF-κB in JB6 P+ cells [89]. CGA inhibited lipopolysaccharide (LPS)-induced inflammatory response in RAW 264.7 cells mediated by decreasing cyclooxygenase (COX-2) at protein and mRNA levels and decreased the secretion of prostaglandin E2 (PGE2) [90]. Chlorogenic acid also inhibited LPS induced inflammation of liver in mice and prevented the mRNA expression of toll-like receptor 4 (TLR4), TNF-α and NF-κB p65 subunit [91].

Effect of hydroxycinnamic acid derivatives on lipid and fat metabolism

Elevated plasma concentrations of total cholesterol (TC) and low density lipoprotein (LDL) cholesterol (and/or reduced high-density lipoprotein [HDL]) are commonly seen in dyslipidemia and strongly associated with cardiovascular disease, peripheral vascular disease and stroke [92]. High fat diet feeding in laboratory animals showed dyslipidemic condition similar to human dyslipidemia. Several plant based compounds e.g., plant stanols and sterols, tea-based catechins and theaflavins showed improvement in lowering plasma lipid profiles; however the clinical efficacy of many of these substances are not well studied [93]. Most of the hydroxycinnamic acid derivatives are effective against fat deposition and lowered plasma lipid profile and increases fat metabolism in liver (Table 1). Polyphenol rich red wine improved plasma lipid profiles by increasing HDL cholesterol levels, improve LDL oxidation [94] and improved the antioxidant status by reducing the oxidative stress in patients [95].

Table 1 Lipid lowering effect of hydroxycinnamic acid derivatives

Cinnamic acid derivative supplementation lowered the plasma and liver triglycerides and cholesterol concentrations in high cholesterol fed rats [96]. Ferulic acid supplementation also lowered plasma lipid and cholesterol concentrations in various model of dyslipidemia such as C57BL/6 mice fed with high fat diet [97], Golden syrian hamsters fed with chow-based hypercholesterolemic diet [98], stroke-prone spontaneously hypertensive rats [99], apolipoprotein E-deficient (apo E−/−) mice fed Western diet [100, 101] and in streptozotocin induced diabetes rats [102]. Chlorogenic acid infusion in diabetic Zucker rats lowered the fasting plasma cholesterol and triacylglycerol concentrations significantly [103]. Chlorogenic acid also lowered the plasma cholesterol in ICR mice fed with high fat diet [15] and lipid, free fatty acids and triglycerides in Streptozotocin (STZ)–nicotinamide (NA)-induced type 2 diabetic rats [104].

Cholesterol lowering effect is attributed to the inhibition of the cholesterol synthesis and utilization of the free fatty acids in liver. HMG-CoA reductase is the rate regulating enzymes found in liver which is responsible for the cholesterol biosynthesis. Several statins selectively inhibited the HMG-CoA reductase in liver and lowered plasma cholesterol in hyperlipidemic patients [105, 106]. Hepatic ACAT is other type of enzymes that increased the utilization of fatty acid for cholesterol biosynthesis. Cinnamic acid derivatives such as cinnamic acid, ferulic acid, chlorgenic acid reduced the HMG-CoA reductase and ACAT activity in experimental animals [96, 101, 15, 104]. Ferulic acid decreased hepatic acyl-coenzyme A: cholesterol acyltransferase activity [100, 101] and down regulates the genes involved in lipid metabolism [99]. Moreover, Chlorgenic acid increased beta-oxidation and lypolitic lipase activity in diabetic animal [15, 104].

Effect of hydroxycinnamic acid derivatives on body weight and obesity

Hydroxycinnamic acids are also effective against body weight gain, fat deposition and dysfunction of the adipocytes due to high fat diet feeding in animal model (Table 2). Adipocyte proliferation and differentiation plays critical role on adipose tissue deposition and dysfunction. 3 T3-L1 preadipocytes are excellent cell lines for studying the anti-obesity effect of various therapeutic agents. Addition of phenolic acids to the growth medium decreased the cell population of 3 T3-L1 preadipocytes in vitro [107]. Chlorogenic acid, o-coumaric acid, and m-coumaric acid caused cell cycle arrest in the G1 phase in 3 T3-L1 preadipocytes [107]. Ferulic acid prevented the body weight gain in high fat diet fed mice and decreased the plasma and liver lipids, triglycerides and total cholesterol [97]. Ferulic acid also decreased the activity of hepatic lipogenic enzymes, such as G6PD, ME, and FAS which are responsible for the cholesterol and fatty acid synthesis [97]. Chlorogenic acid showed anti-obesity effect on mice fed with a high fat diet [15]. Chlorogenic acid also lowered the visceral fat mass and plasma leptin and insulin levels compared to the high-fat control group [15]. Caffeic acid and chlorogenic acid significantly inhibited fatty acid synthase, 3-hydroxy-3-methylglutaryl CoA reductase and acyl-CoA:cholesterol acyltransferase activities, while they increased fatty acid β-oxidation activity and peroxisome proliferator-activated receptors α expression in the liver compared to the high-fat group [15].

Table 2 Effect of hydroxycinnamic acid derivatives on obesity and adipocyte dysfunction

Effect of hydroxycinnamic acid derivatives on diabetes and insulin resistance

Hyperglycemia and insulin resistance are commonly seen in obesity [108–110]. Polyphenolic compounds showed prevention of metabolic disorder associated with hyperglycemia and diabetes. The mechanisms behind these benefits have multiple targets. Some molecules prevented the beta cell destruction in pancreas thereby increasing the insulin secretion. Others include inhibition of carbohydrate digestive enzymes, increased glycogen synthesis, increased glucose uptake in muscle tissues and adipocytes by phosphorylation of AMPK and increased GLUT4 content as well as increasing glucose metabolism. Hydroxycinnamic acid derivatives also showed considerable hypoglycaemic activities in experimental condition (Table 3). Cinnamic acid improved glucose intolerance and insulin resistance in STZ induced diabetic rats [13]. Cinnamic acid also increased the expression of glycogen synthase, whereas the expression of glycogen synthase kinase and phosphorylation of glycogen synthase at Ser641 in TNF-α-treated insulin-resistant mouse hepatocytes was decreased [111]. Rice bran fraction and ferulic acid reduced the blood glucose concentrations and increased the insulin in plasma of diabetic C57BL/KsJ db/db mice [112]. Glucose lowering effect by ferulic acid was also seen in KK-Ay mice [14] and STZ induced diabetic mice [14]. Ferulic acid also increased glucokinase activity [112] and decreased glucose-6-phosphatase (G6pase) and phosphoenolpyruvate carboxykinase (PEPCK) activities in liver [113]. Moreover, ferulic acid prevented lipid peroxidation and improved the antioxidant enzymes such as glutathione peroxidase (GPx), superoxide dismutase (SOD) and catalase (CAT) [114].

Table 3 Effect of hydroxycinnamic acid derivatives on diabetes

Caffeic acid has been studied extensively in experimental diabetes and related complications. Caffeic acid lowered blood glucose level in C57BL/KsJ-db/db mice [115] and Streptozotocin (STZ)-induced diabetic rats [116]. Caffeic acid also improved insulin level in plasma of male Balb/cA mice [11] and improved glucose intolerance in high fat diet fed male mice [117]. Caffeic acid improved insulin resistance by promoting insulin receptor tyrosyl phosphorylation, up-regulate the expression of insulin signal associated proteins, including insulin receptor, phosphatidylinositol-3 kinase, glycogen synthase, and glucose transporter-2, increase the uptake of glucose in tumor necrosis factor-α induced insulin resistant mouse liver FL83B cells [118]. Other studies showed that caffeic acid decreased the inflammatory cytokines [119] and reduced the induction of the inflammatory pathway, c-jun-N- terminal kinase, the nuclear factor kappa B, and cyclooxygenase-2 expression [11]. Furthermore, Caffeic acid increased increased phosphorylation of AMPKs and increased glucose uptake and GLUT4 content in L6-GLUT4myc cells [120]. Chlorogenic acid also follows the similar mechanism for improving insulin resistance and diabetes. Chlorogenic acid stimulates glucose transport in skeletal muscle via the GLUT 4 translocation and phosphorylation of AMPK and Akt in db/db mice [121].

Conclusion

Recent research has provided the scientific benefit of these phenolic acids and confirmed the important role of phenolic acids in the prevention and treatment of obesity, diabetes and associated disorders. Phenolic acids could favourably affect most of the leading aspects of obesity including diabetes, including insulin resistance, hyperglycemia, hyperlipidemia, and adepocyte dysfunction and inflammation (Fig. 3). Despite the potential benefits of these natural products in preclinical studies, scanty literatures have been found on any beneficial effect from clinical trials of phenolic acids so far. Studies are thus required in humans to confirm the potential benefit of phenolic acids in limiting obesity and other associated disorders. Furthermore, multiple approaches are also needed to overcome limited solubility and poor bioavailability of phenolic acids. These include synthesis of phenolic acids derivatives and development of novel drug delivery system and formulations such as nanoparticles, liposomal encapsulation, emulsions, and sustained released tablets. Therefore, enhanced bioavailability and convinced clinical trial results of phenolic acids could bring these promising natural products to the forefront of therapeutic agents for obesity.

Fig. 3
figure 3

Hypothetical representation of fat metabolism in response to hydroxycinnamic acid derivatives

Abbreviations

AGEs:

advanced glycation end products

AMPK:

AMP-activated protein kinase

AP-1:

activator protein-1 DNA binding

CPT-1:

carnitin palmitoyl transferase-1.

EGCG:

epigalocatechin gallate

ERK:

extracellular signal-regulated protein kinase

GIP:

glucose-dependent insulinotropic polypeptide

GLUT:

glucose transporter

MAPK:

mitogen-activated protein kinases

MMP-2:

matrix metalloproteinase-2

NF-κB:

nuclear factor kappa-B

Nrf2:

nuclear factor erythroid 2 related factor 2

PEPCK:

glucose-6-phosphatase and phosphoenolpyruvate carboxykinase

PGI2:

increased prostacyclin I2

PI3:

phosphoinositide 3 kinase/protein kinase B

PPARα:

peroxisome proliferator-activated receptor alpha

ROS:

reactive oxygen species

SGLT:

sodium-dependent glucose transporter

TGF-β:

transforming growth factor-β

TXA2:

thromboxane A2

UCP-2:

uncoupling protein 2

VEGF:

vascular endothelial growth factor

References

  1. Hu FB. Globalization of diabetes: the role of diet, lifestyle, and genes. Diabetes Care. 2011;34(6):1249–57.

    Article  Google Scholar 

  2. Gupta N, Goel K, Shah P, Misra A. Childhood obesity in developing countries: epidemiology, determinants, and prevention. Endocr Rev. 2012;33(1):48–70.

    Article  CAS  Google Scholar 

  3. Popkin BM, Adair LS, Ng SW. Global nutrition transition and the pandemic of obesity in developing countries. Nutr Rev. 2012;70(1):3–21.

    Article  Google Scholar 

  4. Bruno G, Landi A. Epidemiology and costs of diabetes. Transplant Proc. 2011;43(1):327–9.

    Article  CAS  Google Scholar 

  5. Marinou K, Tousoulis D, Antonopoulos AS, Stefanadi E, Stefanadis C. Obesity and cardiovascular disease: from pathophysiology to risk stratification. Int J Cardiol. 2010;138(1):3–8.

    Article  Google Scholar 

  6. Berrington de Gonzalez A, Hartge P, Cerhan JR, Flint AJ, Hannan L, MacInnis RJ, Moore SC, Tobias GS, Anton-Culver H, Freeman LB et al. Body-mass index and mortality among 1.46 million white adults. New Engl J Med. 2010;363(23):2211–9.

    Article  CAS  Google Scholar 

  7. Lazarou C, Panagiotakos D, Matalas AL. The role of diet in prevention and management of type 2 diabetes: implications for public health. Crit Rev Food Sci Nutr. 2012;52(5):382–9.

    Article  CAS  Google Scholar 

  8. Pistollato F, Battino M. Role of plant-based diets in the prevention and regression of metabolic syndrome and neurodegenerative diseases. Trends Food Sci Technol. 2014.

  9. Rajarathnam S, Shashirakha MN, Mallikarjuna SE. Status of bioactive compounds in foods, with focus on fruits and vegetables. Crit Rev Food Sci Nutr. 2013;55:1324–39. doi:10.1080/10408398.2012.692736.

    Google Scholar 

  10. Arranz S, Chiva-Blanch G, Valderas-Martínez P, Medina-Remón A, Lamuela-Raventós RM, Estruch R. Wine, beer, alcohol and polyphenols on cardiovascular disease and cancer. Nutrients. 2012;4(7):759–81.

    Article  CAS  Google Scholar 

  11. Cy C, Mc M, Kc C, Mc Y. Anti-glycative and anti-inflammatory effects of caffeic acid and ellagic acid in kidney of diabetic mice. Mol Nutr Food Res. 2010;54(3):388–95.

    Article  CAS  Google Scholar 

  12. Hsu CL, Wu CH, Huang SL, Yen GC. Phenolic compounds rutin and o-coumaric acid ameliorate obesity induced by high-fat diet in rats. J Agric Food Chem. 2009;57(2):425–31.

    Article  CAS  Google Scholar 

  13. Kasetti RB, Nabi SA, Swapna S, Apparao C. Cinnamic acid as one of the antidiabetic active principle(s) from the seeds of Syzygium alternifolium. Food Chem Toxicol. 2012;50(5):1425–31.

    Article  CAS  Google Scholar 

  14. Ohnishi M, Matuo T, Tsuno T, Hosoda A, Nomura E, Taniguchi H, Sasaki H, Morishita H:. Antioxidant activity and hypoglycemic effect of ferulic acid in STZ-induced diabetic mice and KK-Ay mice. BioFactors. 2004;21(1–4):315–9.

    Article  CAS  Google Scholar 

  15. Cho AS, Jeon SM, Kim MJ, Yeo J, Seo KI, Choi MS, Lee MK. Chlorogenic acid exhibits anti-obesity property and improves lipid metabolism in high-fat diet-induced-obese mice. Food Chem Toxicol. 2010;48(3):937–43.

    Article  CAS  Google Scholar 

  16. Chao PC, Hsu CC, Yin MC. Anti-inflammatory and anti-coagulatory activities of caffeic acid and ellagic acid in cardiac tissue of diabetic mice. Nutr Metab (Lond). 2009;6:33.

    Article  CAS  Google Scholar 

  17. Organization: WHO: Obesity and overweight. Fact sheet No. 311, updated January 2015. http://www.who.int/mediacentre/factsheets/fs311/en/webcite. Accessed 29 August 2015.

  18. Herman CP, Polivy J. Self-regulation and the obesity epidemic. Soc Issues Pol Rev. 2011;5(1):37–69.

    Article  Google Scholar 

  19. Wang Y, Lim H. The global childhood obesity epidemic and the association between socio-economic status and childhood obesity. Int Rev Psychiatry. 2012;24(3):176–88.

    Article  Google Scholar 

  20. Racette SB, Deusinger SS, Deusinger RH. Obesity: overview of prevalence, etiology, and treatment. Phys Ther. 2003;83(3):276–88.

    Google Scholar 

  21. Choudhury M, Jonscher KR, Friedman JE. Reduced mitochondrial function in obesity-associated fatty liver: SIRT3 takes on the fat. Aging. 2011;3(2):175–8.

    Article  CAS  Google Scholar 

  22. Nicolson GL. Metabolic syndrome and mitochondrial function: molecular replacement and antioxidant supplements to prevent membrane peroxidation and restore mitochondrial function. J Cell Biochem. 2007;100(6):1352–69.

    Article  CAS  Google Scholar 

  23. Nisoli E, Clementi E, Carruba MO, Moncada S. Defective mitochondrial biogenesis. Circul Res. 2007;100(6):795–806.

    Article  CAS  Google Scholar 

  24. Pagel-Langenickel I, Bao J, Pang L, Sack MN. The role of mitochondria in the pathophysiology of skeletal muscle insulin resistance. Endocr Rev. 2010;31(1):25–51.

    Article  CAS  Google Scholar 

  25. Seifert EL, Estey C, Xuan JY, Harper ME. Electron transport chain-dependent and -independent mechanisms of mitochondrial H2O2 emission during long-chain fatty acid oxidation. J Biol Chem. 2010;285(8):5748–58.

    Article  CAS  Google Scholar 

  26. Hey-Mogensen M, Jeppesen J, Madsen K, Kiens B, Franch J. Obesity augments the age-induced increase in mitochondrial capacity for H2O2 release in Zucker fatty rats. Acta Physiol. 2012;204(3):354–61.

    Article  CAS  Google Scholar 

  27. James AM, Collins Y, Logan A, Murphy MP. Mitochondrial oxidative stress and the metabolic syndrome. Trends Endocrinol Metab. 2012;23(9):429–34.

    Article  CAS  Google Scholar 

  28. Puigserver P, Spiegelman BM. Peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α): transcriptional coactivator and metabolic regulator. Endocr Rev. 2003;24(1):78–90.

    Article  CAS  Google Scholar 

  29. Vega RB, Huss JM, Kelly DP. The coactivator PGC-1 cooperates with peroxisome proliferator-activated receptor alpha in transcriptional control of nuclear genes encoding mitochondrial fatty acid oxidation enzymes. Mol Cell Biol. 2000;20(5):1868–76.

    Article  CAS  Google Scholar 

  30. Hock MB, Kralli A. Transcriptional control of mitochondrial biogenesis and function. Annu Rev Physiol. 2009;71:177–203.

    Article  CAS  Google Scholar 

  31. Zhang BB, Zhou G, Li C. AMPK: an emerging drug target for diabetes and the metabolic syndrome. Cell Metab. 2009;9(5):407–16.

    Article  CAS  Google Scholar 

  32. Cantó C, Auwerx J. PGC-1[alpha], SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr Opin Lipidol. 2009;20(2):98–105. doi:10.1097/MOL.1090b1013e328328d328320a328324.

    Article  CAS  Google Scholar 

  33. Hardie DG. AMPK: a key regulator of energy balance in the single cell and the whole organism. Int J Obes. 2008;32(S4):S7–12.

    Article  CAS  Google Scholar 

  34. Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: Ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005;1(1):15–25.

    Article  CAS  Google Scholar 

  35. Zong H, Ren JM, Young LH, Pypaert M, Mu J, Birnbaum MJ, Shulman GI. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proc Natl Acad Sci. 2002;99(25):15983–7.

    Article  CAS  Google Scholar 

  36. Garcia-Roves PM, Osler ME, Holmström MH, Zierath JR. Gain-of-function R225Q mutation in AMP-activated protein kinase γ3 subunit increases mitochondrial biogenesis in glycolytic skeletal muscle. J Biol Chem. 2008;283(51):35724–34.

    Article  CAS  Google Scholar 

  37. Long Y, Barnes B, Mahlapuu M, Steiler T, Martinsson S, Leng Y, Wallberg-Henriksson H, Andersson L, Zierath J. Role of AMP-activated protein kinase in the coordinated expression of genes controlling glucose and lipid metabolism in mouse white skeletal muscle. Diabetologia. 2005;48(11):2354–64.

    Article  CAS  Google Scholar 

  38. Fryer LGD, Parbu-Patel A, Carling D. The anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathways. J Biol Chem. 2002;277(28):25226–32.

    Article  CAS  Google Scholar 

  39. Brunmair B, Staniek K, Gras F, Scharf N, Althaym A, Clara R, Roden M, Gnaiger E, Nohl H, Waldhäusl W, et al. Thiazolidinediones, like metformin, inhibit respiratory complex I. Diabetes. 2004;53(4):1052–9.

    Article  CAS  Google Scholar 

  40. Cool B, Zinker B, Chiou W, Kifle L, Cao N, Perham M, Dickinson R, Adler A, Gagne G, Iyengar R, et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006;3(6):403–16.

    Article  CAS  Google Scholar 

  41. Suwa M, Nakano H, Kumagai S. Effects of chronic AICAR treatment on fiber composition, enzyme activity, UCP3, and PGC-1 in rat muscles. J Appl Physiol. 2003;95(3):960–8.

    Article  CAS  Google Scholar 

  42. Lee WJ, Kim M, Park H-S, Kim HS, Jeon MJ, Oh KS, Koh EH, Won JC, Kim MS, Oh GT, et al. AMPK activation increases fatty acid oxidation in skeletal muscle by activating PPARα and PGC-1. Biochem Biophys Res Commun. 2006;340(1):291–5.

    Article  CAS  Google Scholar 

  43. Chawla A, Schwarz EJ, Dimaculangan DD, Lazar MA. Peroxisome proliferator-activated receptor (PPAR) gamma: adipose-predominant expression and induction early in adipocyte differentiation. Endocrinology. 1994;135(2):798–800.

    CAS  Google Scholar 

  44. Tontonoz P, Hu E, Graves RA, Budavari AI, Spiegelman BM. mPPAR gamma 2: tissue-specific regulator of an adipocyte enhancer. Genes Dev. 1994;8(10):1224–34.

    Article  CAS  Google Scholar 

  45. Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, Laville M, Fruchart JC, Deeb S, et al. The organization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem. 1997;272(30):18779–89.

    Article  CAS  Google Scholar 

  46. Cock TA, Houten SM, Auwerx J. Peroxisome proliferator-activated receptor-[gamma]: too much of a good thing causes harm. EMBO Rep. 2004;5(2):142–7.

    Article  CAS  Google Scholar 

  47. Miles PD, Barak Y, He W, Evans RM, Olefsky JM. Improved insulin-sensitivity in mice heterozygous for PPAR-gamma deficiency. J Clin Invest. 2000;105(3):287–92.

    Article  CAS  Google Scholar 

  48. Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T, et al. PPAR3 mediates high-Fat diet induced adipocyte hypertrophy and insulin resistance. Mol Cell. 1999;4(4):597–609.

    Article  CAS  Google Scholar 

  49. Szanto A, Nagy L. The many faces of PPARgamma: anti-inflammatory by any means? Immunobiology. 2008;213(9–10):789–803.

    Article  CAS  Google Scholar 

  50. Lehrke M, Lazar MA. The many faces of PPARgamma. Cell. 2005;123(6):993–9.

    Article  CAS  Google Scholar 

  51. He W, Barak Y, Hevener A, Olson P, Liao D, Le J, Nelson M, Ong E, Olefsky JM, Evans RM. Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle. Proc Natl Acad Sci U S A. 2003;100(26):15712–7.

    Article  CAS  Google Scholar 

  52. Hevener AL, He W, Barak Y, Le J, Bandyopadhyay G, Olson P, Wilkes J, Evans RM, Olefsky J. Muscle-specific Pparg deletion causes insulin resistance. Nat Med. 2003;9(12):1491–7.

    Article  CAS  Google Scholar 

  53. Norris AW, Chen L, Fisher SJ, Szanto I, Ristow M, Jozsi AC, Hirshman MF, Rosen ED, Goodyear LJ, Gonzalez FJ, et al. Muscle-specific PPARgamma-deficient mice develop increased adiposity and insulin resistance but respond to thiazolidinediones. J Clin Invest. 2003;112(4):608–18.

    Article  CAS  Google Scholar 

  54. Matsusue K, Haluzik M, Lambert G, Yim SH, Gavrilova O, Ward JM, Brewer B, Jr., Reitman ML, Gonzalez FJ. Liver-specific disruption of PPARgamma in leptin-deficient mice improves fatty liver but aggravates diabetic phenotypes. J Clin Invest. 2003;111(5):737–47.

    Article  CAS  Google Scholar 

  55. Rangwala SM, Rhoades B, Shapiro JS, Rich AS, Kim JK, Shulman GI, Kaestner KH, Lazar MA. Genetic modulation of PPARgamma phosphorylation regulates insulin sensitivity. Dev Cell. 2003;5(4):657–63.

    Article  CAS  Google Scholar 

  56. Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor γ (PPARγ). J Biol Chem. 1995;270(22):12953–6.

    Article  CAS  Google Scholar 

  57. Fonseca V. Effect of thiazolidinediones on body weight in patients with diabetes mellitus. Am J Med. 2003;115(8):42–8.

    Article  Google Scholar 

  58. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell. 1998;92(6):829–39.

    Article  CAS  Google Scholar 

  59. Liang H, Ward WF. PGC-1α: a key regulator of energy metabolism. Adv Physiol Educ. 2006;30(4):145–51.

    Article  Google Scholar 

  60. Fernandez-Marcos PJ, Auwerx J. Regulation of PGC-1α, a nodal regulator of mitochondrial biogenesis. Am J Clin Nutr. 2011;93(4):884S–90.

    Article  CAS  Google Scholar 

  61. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1α. Cell. 2006;127(6):1109–22.

    Article  CAS  Google Scholar 

  62. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444(7117):337–42.

    Article  CAS  Google Scholar 

  63. de Luca C, Olefsky JM. Inflammation and insulin resistance. FEBS Lett. 2008;582(1):97–105.

    Article  CAS  Google Scholar 

  64. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–46.

    Article  CAS  Google Scholar 

  65. Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. J Clin Invest. 2011;121(6):2111–7.

    Article  CAS  Google Scholar 

  66. Maury E, Brichard SM. Adipokine dysregulation, adipose tissue inflammation and metabolic syndrome. Mol Cell Endocrinol. 2010;314(1):1–16.

    Article  CAS  Google Scholar 

  67. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. 1995;95(5):2409–15.

    Article  CAS  Google Scholar 

  68. Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW, 2nd, DeFuria J, Jick Z, Greenberg AS, Obin MS. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56(12):2910–8.

    Article  CAS  Google Scholar 

  69. Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K, Furukawa S, Tochino Y, Komuro R, Matsuda M, et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes. 2007;56(4):901–11.

    Article  CAS  Google Scholar 

  70. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, Tuncman G, Gorgun C, Glimcher LH, Hotamisligil GS. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306(5695):457–61.

    Article  CAS  Google Scholar 

  71. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante Jr AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808.

    Article  CAS  Google Scholar 

  72. Rodríguez-Hernández H, Simental-Mendía LE, Rodríguez-Ramírez G, Reyes-Romero MA. Obesity and inflammation: epidemiology, risk factors, and markers of inflammation. Int J Endocrinol. 2013;2013:11. doi:10.1155/2013/678159.

    Article  Google Scholar 

  73. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–30.

    Article  CAS  Google Scholar 

  74. El-Seedi HR, El-Said AM, Khalifa SA, Goransson U, Bohlin L, Borg-Karlson AK, Verpoorte R. Biosynthesis, natural sources, dietary intake, pharmacokinetic properties, and biological activities of hydroxycinnamic acids. J Agric Food Chem. 2012;60(44):10877–95.

    Article  CAS  Google Scholar 

  75. Zhao Z, Moghadasian MH. Chemistry, natural sources, dietary intake and pharmacokinetic properties of ferulic acid: A review. Food Chem. 2008;109(4):691–702.

    Article  CAS  Google Scholar 

  76. Lafay S, Gil-Izquierdo A. Bioavailability of phenolic acids. Phytochem Rev. 2008;7(2):301–11.

    Article  CAS  Google Scholar 

  77. Konishi Y, Kobayashi S. Transepithelial transport of chlorogenic acid, caffeic acid, and their colonic metabolites in intestinal caco-2 cell monolayers. J Agric Food Chem. 2004;52(9):2518–26.

    Article  CAS  Google Scholar 

  78. Konishi Y, Hitomi Y, Yoshioka E. Intestinal absorption of p-coumaric and gallic acids in rats after oral administration. J Agric Food Chem. 2004;52(9):2527–32.

    Article  CAS  Google Scholar 

  79. Poquet L, Clifford MN, Williamson G. Transport and metabolism of ferulic acid through the colonic epithelium. Drug Metab Disposition. 2008;36(1):190–7.

    Article  CAS  Google Scholar 

  80. Wolffram S, Weber T, Grenacher B, Scharrer E. A Na + −dependent mechanism is involved in mucosal uptake of cinnamic acid across the jejunal brush border in rats. J Nutr. 1995;125(5):1300–8.

    CAS  Google Scholar 

  81. Kim EO, Min KJ, Kwon TK, Um BH, Moreau RA, Choi SW. Anti-inflammatory activity of hydroxycinnamic acid derivatives isolated from corn bran in lipopolysaccharide-stimulated Raw 264.7 macrophages. Food Chem Toxicol. 2012;50(5):1309–16.

    Article  CAS  Google Scholar 

  82. Sakai S, Ochiai H, Nakajima K, Terasawa K. Inhibitory effect of ferulic acid on macrophage inflammatory protein-2 production in a murine macrophage cell line, Raw264.7. Cytokine. 1997;9(4):242–8.

    Article  CAS  Google Scholar 

  83. Hole AS, Grimmer S, Jensen MR, Sahlstrøm S. Synergistic and suppressive effects of dietary phenolic acids and other phytochemicals from cereal extracts on nuclear factor kappa B activity. Food Chem. 2012;133(3):969–77.

    Article  CAS  Google Scholar 

  84. Jung KJ, Go EK, Kim JY, Yu BP, Chung HY. Suppression of age-related renal changes in NF-κB and its target gene expression by dietary ferulate. J Nutr Biochem. 2009;20(5):378–88.

    Article  CAS  Google Scholar 

  85. Pragasam S, Venkatesan V, Rasool M. Immunomodulatory and anti-inflammatory effect of p-coumaric acid, a common dietary polyphenol on experimental inflammation in rats. Inflammation. 2012;1–8.

  86. Yen GC, Chen YC, Chang WT, Hsu CL. Effects of Polyphenolic Compounds on Tumor Necrosis Factor-α (TNF-α)-Induced Changes of Adipokines and Oxidative Stress in 3 T3-L1 Adipocytes. J Agric Food Chem. 2010;59(2):546–51.

    Article  CAS  Google Scholar 

  87. Michaluart P, Masferrer JL, Carothers AM, Subbaramaiah K, Zweifel BS, Koboldt C, Mestre JR, Grunberger D, Sacks PG, Tanabe T, et al. Inhibitory effects of caffeic acid phenethyl ester on the activity and expression of cyclooxygenase-2 in human oral epithelial cells and in a rat model of inflammation. Cancer Res. 1999;59(10):2347–52.

    CAS  Google Scholar 

  88. da Cunha FM, Duma D, Assreuy J, Buzzi FC, Niero R, Campos MM, Calixto JB. Caffeic acid derivatives: in vitro and in vivo anti-inflammatory properties. Free Radic Res. 2004;38(11):1241–53.

    Article  CAS  Google Scholar 

  89. Feng R, Lu Y, Bowman LL, Qian Y, Castranova V, Ding M. Inhibition of activator protein-1, NF-κB, and MAPKs and induction of phase 2 detoxifying enzyme activity by chlorogenic acid. J Biol Chem. 2005;280(30):27888–95.

    Article  CAS  Google Scholar 

  90. Shan J, Fu J, Zhao Z, Kong X, Huang H, Luo L, Yin Z. Chlorogenic acid inhibits lipopolysaccharide-induced cyclooxygenase-2 expression in RAW264.7 cells through suppressing NF-κB and JNK/AP-1 activation. Int Immunopharmacol. 2009;9(9):1042–8.

    Article  CAS  Google Scholar 

  91. Xu Y, Chen J, Yu X, Tao W, Jiang F, Yin Z, Liu C. Protective effects of chlorogenic acid on acute hepatotoxicity induced by lipopolysaccharide in mice. Inflammation Res. 2010;59(10):871–7.

    Article  CAS  Google Scholar 

  92. Ferdowsian HR, Barnard ND. Effects of plant-based diets on plasma lipids. Am J Cardiol. 2009;104(7):947–56.

    Article  CAS  Google Scholar 

  93. McGowan M, Proulx S. Nutritional supplements and serum lipids: Does anything work? Curr Atheroscler Rep. 2009;11(6):470–6.

    Article  CAS  Google Scholar 

  94. Hansen AS, Marckmann P, Dragsted LO, Finne Nielsen IL, Nielsen SE, Gronbaek M. Effect of red wine and red grape extract on blood lipids, haemostatic factors, and other risk factors for cardiovascular disease. Eur J Clin Nutr. 2005;59(3):449–55.

    Article  CAS  Google Scholar 

  95. Pignatelli P, Ghiselli A, Buchetti B, Carnevale R, Natella F, Germano G, Fimognari F, Di Santo S, Lenti L, Violi F. Polyphenols synergistically inhibit oxidative stress in subjects given red and white wine. Atherosclerosis. 2006;188(1):77–83.

    Article  CAS  Google Scholar 

  96. Lee MK, Park YB, Moon SS, Bok SH, Kim DJ, Ha TY, Jeong TS, Jeong KS, Choi MS. Hypocholesterolemic and antioxidant properties of 3-(4-hydroxyl)propanoic acid derivatives in high-cholesterol fed rats. Chem Biol Interact. 2007;170(1):9–19.

    Article  CAS  Google Scholar 

  97. Jin Son M, Rico CW, Hyun Nam S, Young Kang M. Influence of oryzanol and ferulic Acid on the lipid metabolism and antioxidative status in high fat-fed mice. J Clin Biochem Nutr. 2010;46(2):150–6.

    Article  Google Scholar 

  98. Wilson TA, Nicolosi RJ, Woolfrey B, Kritchevsky D. Rice bran oil and oryzanol reduce plasma lipid and lipoprotein cholesterol concentrations and aortic cholesterol ester accumulation to a greater extent than ferulic acid in hypercholesterolemic hamsters. J Nutr Biochem. 2007;18(2):105–12.

    Article  CAS  Google Scholar 

  99. Ardiansyah, Ohsaki Y, Shirakawa H, Koseki T, Komai M. Novel effects of a single administration of ferulic acid on the regulation of blood pressure and the hepatic lipid metabolic profile in stroke-prone spontaneously hypertensive rats. J Agric Food Chem. 2008;56(8):2825–30.

    Article  CAS  Google Scholar 

  100. Kwon EY, Cho YY, Do GM, Kim HJ, Jeon SM, Park YB, Lee MK, Min TS, Choi MS. Actions of ferulic acid and vitamin E on prevention of hypercholesterolemia and atherogenic lesion formation in apolipoprotein E-deficient mice. J Med Food. 2009;12(5):996–1003.

    Article  CAS  Google Scholar 

  101. Kwon EY, Do GM, Cho YY, Park YB, Jeon SM, Choi MS. Anti-atherogenic property of ferulic acid in apolipoprotein E-deficient mice fed Western diet: Comparison with clofibrate. Food Chem Toxicol. 2010;48(8–9):2298–303.

    Article  CAS  Google Scholar 

  102. Sri Balasubashini M, Rukkumani R, Menon VP. Protective effects of ferulic acid on hyperlipidemic diabetic rats. Acta Diabetol. 2003;40(3):118–22.

    Article  CAS  Google Scholar 

  103. de Sotillo DV R, Hadley M. Chlorogenic acid modifies plasma and liver concentrations of: cholesterol, triacylglycerol, and minerals in (fa/fa) Zucker rats. J Nutr Biochem. 2002;13(12):717–26.

    Article  Google Scholar 

  104. Karthikesan K, Pari L, Menon VP. Antihyperlipidemic effect of chlorogenic acid and tetrahydrocurcumin in rats subjected to diabetogenic agents. Chem Biol Interact. 2010;188(3):643–50.

    Article  CAS  Google Scholar 

  105. Gazzerro P, Proto MC, Gangemi G, Malfitano AM, Ciaglia E, Pisanti S, Santoro A, Laezza C, Bifulco M. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev. 2012;64(1):102–46.

    Article  CAS  Google Scholar 

  106. Pahan K. Lipid-lowering drugs. Cell Mol Life Sci. 2006;63(10):1165–78.

    Article  CAS  Google Scholar 

  107. Hsu CL, Huang SL, Yen GC. Inhibitory effect of phenolic acids on the proliferation of 3 T3-L1 preadipocytes in relation to their antioxidant activity. J Agric Food Chem. 2006;54(12):4191–7.

    Article  CAS  Google Scholar 

  108. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840–6.

    Article  CAS  Google Scholar 

  109. Qatanani M, Lazar MA. Mechanisms of obesity-associated insulin resistance: many choices on the menu. Genes Dev. 2007;21(12):1443–55.

    Article  CAS  Google Scholar 

  110. Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest. 2000;106(4):473–81.

    Article  CAS  Google Scholar 

  111. Huang DW, Shen SC. Caffeic acid and cinnamic acid ameliorate glucose metabolism via modulating glycogenesis and gluconeogenesis in insulin-resistant mouse hepatocytes. J Funct Foods. 2012;4(1):358–66.

    Article  CAS  Google Scholar 

  112. Jung EH, Kim SR, Hwang IK, Ha TY. Hypoglycemic effects of a phenolic acid fraction of rice bran and ferulic acid in C57BL/KsJ-db/db mice. J Agric Food Chem. 2007;55(24):9800–4.

    Article  CAS  Google Scholar 

  113. Son MJ, Rico CW, Nam SH, Kang MY. Effect of oryzanol and ferulic acid on the glucose metabolism of mice Fed with a high-Fat diet. J Food Sci. 2011;76(1):H7–10.

    Article  CAS  Google Scholar 

  114. Balasubashini MS, Rukkumani R, Viswanathan P, Menon VP. Ferulic acid alleviates lipid peroxidation in diabetic rats. Phytother Res. 2004;18(4):310–4.

    Article  CAS  Google Scholar 

  115. Jung UJ, Lee MK, Park YB, Jeon SM, Choi MS. Antihyperglycemic and antioxidant properties of caffeic acid in db/db mice. J Pharmacol Exp Ther. 2006;318(2):476–83.

    Article  CAS  Google Scholar 

  116. Celik S, Erdogan S, Tuzcu M. Caffeic acid phenethyl ester (CAPE) exhibits significant potential as an antidiabetic and liver-protective agent in streptozotocin-induced diabetic rats. Pharmacol Res. 2009;60(4):270–6.

    Article  CAS  Google Scholar 

  117. Bezerra RMN, Veiga LF, Caetano AC, Rosalen PL, Amaral MEC, Palanch AC, de Alencar SM. Caffeic acid phenethyl ester reduces the activation of the nuclear factor κB pathway by high-fat diet-induced obesity in mice. Metabolism. 2012;61(11):1606–14.

    Article  CAS  Google Scholar 

  118. Huang DW, Shen SC, Wu JSB. Effects of caffeic acid and cinnamic acid on glucose uptake in insulin-resistant mouse hepatocytes. J Agric Food Chem. 2009;57(17):7687–92.

    Article  CAS  Google Scholar 

  119. Weng YC, Chuang ST, Lin YC, Chuang CF, Chi TC, Chiu HL, Kuo YH, Su MJ. Caffeic acid phenylethyl amide protects against the metabolic consequences in diabetes mellitus induced by diet and streptozocin. Evid Based Complement Alternat Med. 2012;2012:12.

    Google Scholar 

  120. Eid HM, Thong F, Sweeney G, Haddad PS. Caffeic acid methyl and ethyl esters induce the translocation of glucose transporter GLUT4 in cultured skeletal muscle cells. Planta Med. 2012;78(11):D121.

    Google Scholar 

  121. Ong KW, Hsu A, Tan BK. Chlorogenic acid stimulates glucose transport in skeletal muscle via AMPK activation: a contributor to the beneficial effects of coffee on diabetes. PLoS One. 2012;7(3), e32718.

    Article  CAS  Google Scholar 

  122. Mnafgui K, Derbali A, Sayadi S, Gharsallah N, Elfeki A, Allouche N. Anti-obesity and cardioprotective effects of cinnamic acid in high fat diet- induced obese rats. J Food Sci Technol. 2014;52(7):4369–77.

    Article  CAS  Google Scholar 

  123. Kopp C, Singh SP, Regenhard P, Muller U, Sauerwein H, Mielenz M. Trans-cinnamic acid increases adiponectin and the phosphorylation of AMP-activated protein kinase through G-protein-coupled receptor signaling in 3 T3-L1 adipocytes. Int J Mol Sci. 2014;15(2):2906–15.

    Article  CAS  Google Scholar 

  124. Hsu CL, Yen GC. Effects of flavonoids and phenolic acids on the inhibition of adipogenesis in 3 T3-L1 adipocytes. J Agric Food Chem. 2007;55(21):8404–10.

    Article  CAS  Google Scholar 

  125. Yen GC, Chen YC, Chang WT, Hsu CL. Effects of polyphenolic compounds on tumor necrosis factor-alpha (TNF-alpha)-induced changes of adipokines and oxidative stress in 3 T3-L1 adipocytes. J Agric Food Chem. 2011;59(2):546–51.

    Article  CAS  Google Scholar 

  126. Juman S, Yasui N, Okuda H, Ueda A, Negishi H, Miki T, Ikeda K. Caffeic acid phenethyl ester suppresses the production of adipocytokines, leptin, tumor necrosis factor -alpha and resistin, during differentiation to adipocytes in 3 T3-L1 cells. Biol Pharm Bull. 2011;34(4):490–4.

    Article  CAS  Google Scholar 

  127. Li S-Y, Chang CQ, Ma FY, Yu CL. Modulating effects of chlorogenic acid on lipids and glucose metabolism and expression of hepatic peroxisome proliferator-activated receptor-α in golden hamsters Fed on high Fat diet. Biomed Environ Sci. 2009;22(2):122–9.

    Article  CAS  Google Scholar 

  128. Wan CW, Wong CN, Pin WK, Wong MH, Kwok CY, Chan RY, Yu PH, Chan SW. Chlorogenic acid exhibits cholesterol lowering and fatty liver attenuating properties by Up-regulating the gene expression of PPAR-alpha in hypercholesterolemic rats induced with a high-cholesterol diet. Phytother Res. 2012;27(4):545–51.

    Article  CAS  Google Scholar 

  129. Ardiansyah, Shirakawa H, KosekiT T, Hashizume K, Komai M. The Driselase-treated fraction of rice bran is a more effective dietary factor to improve hypertension, glucose and lipid metabolism in stroke-prone spontaneously hypertensive rats compared to ferulic acid. Br J Nutr. 2007;97(1):67–76.

    Article  CAS  Google Scholar 

  130. Narasimhan A, Chinnaiyan M, Karundevi B. Ferulic acid exerts its antidiabetic effect by modulating insulin-signalling molecules in the liver of high-fat diet and fructose-induced type-2 diabetic adult male rat. Appl Physiol Nutr Metab. 2015;40(8):769–81.

    Article  CAS  Google Scholar 

  131. Narasimhan A, Chinnaiyan M, Karundevi B. Ferulic acid regulates hepatic GLUT2 gene expression in high fat and fructose-induced type-2 diabetic adult male rat. Eur J Pharmacol. 2015;761:391–7.

    Article  CAS  Google Scholar 

  132. Yilmaz HR, Uz E, Yucel N, Altuntas I, Ozcelik N. Protective effect of caffeic acid phenethyl ester (CAPE) on lipid peroxidation and antioxidant enzymes in diabetic rat liver. J Biochem Mol Toxicol. 2004;18(4):234–8.

    Article  CAS  Google Scholar 

  133. Tsuda S, Egawa T, Ma X, Oshima R, Kurogi E, Hayashi T. Coffee polyphenol caffeic acid but not chlorogenic acid increases 5’AMP-activated protein kinase and insulin-independent glucose transport in rat skeletal muscle. J Nutr Biochem. 2012;23(11):1403–9.

    Article  CAS  Google Scholar 

  134. Tunnicliffe JM, Eller LK, Reimer RA, Hittel DS, Shearer J. Chlorogenic acid differentially affects postprandial glucose and glucose-dependent insulinotropic polypeptide response in rats. Appl Physiol Nutr Metab. 2011;36(5):650–9.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M Obayed Ullah.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

All authors read and approved the final manuscript.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alam, M.A., Subhan, N., Hossain, H. et al. Hydroxycinnamic acid derivatives: a potential class of natural compounds for the management of lipid metabolism and obesity. Nutr Metab (Lond) 13, 27 (2016). https://doi.org/10.1186/s12986-016-0080-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12986-016-0080-3

Keywords