Acessibilidade / Reportar erro

Ferulic acid lowers body weight and visceral fat accumulation via modulation of enzymatic, hormonal and inflammatory changes in a mouse model of high-fat diet-induced obesity

Abstract

Previous studies have reported on the glucose and lipid-lowering effects of ferulic acid (FA) but its anti-obesity potential has not yet been firmly established. This study investigated the possible anti-obesitogenic effects of FA in mice fed a high-fat diet (HFD) for 15 weeks. To assess the antiobesity potential of FA, 32 male Swiss mice, weighing 20–25 g (n=6–8 per group) were fed a normal diet (ND) or HFD, treated orally or not with either FA (10 mg/kg) or sibutramine (10 mg/kg) for 15 weeks and at the end of this period, the body weights of animals, visceral fat accumulation, plasma levels of glucose and insulin hormone, amylase and lipase activities, the satiety hormones ghrelin and leptin, and tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCH-1) were analyzed. Results revealed that FA could effectively suppress the HFD-associated increase in visceral fat accumulation, adipocyte size and body weight gain, similar to sibutramine, the positive control. FA also significantly (P<0.05) decreased the HFD-induced elevations in serum lipid profiles, amylase and lipase activities, and the levels of blood glucose and insulin hormone. The markedly elevated leptin and decreased ghrelin levels seen in HFD-fed control mice were significantly (P<0.05) reversed by FA treatment, almost reaching the values seen in ND-fed mice. Furthermore, FA demonstrated significant (P<0.05) inhibition of serum levels of inflammatory mediators TNF-α, and MCH-1. These results suggest that FA could be beneficial in lowering the risk of HFD-induced obesity via modulation of enzymatic, hormonal and inflammatory responses.

Ferulic acid; Anti-obesity; High-fat diet; Leptin; Lipase; Tumor necrosis factor-α


Introduction

The increasing trend in the prevalence of obesity has become a global concern, causing severe burden on health care systems (11. Caveney E, Caveney BJ, Somaratne R, Turner JR, Gourgiotis L. Pharmaceutical interventions for obesity: a public health perspective. Diabetes Obes Metab 2011; 13: 490–497, doi: 10.1111/j.1463-1326.2010.01353.x.
https://doi.org/10.1111/j.1463-1326.2010...
). Characterized by the accumulation of excess adipose tissue, obesity is related to cardiovascular disease, insulin resistance, and metabolic syndrome (22. Ohman MK, Wright AP, Wickenheiser KJ, Luo W, Eitzman DT. Visceral adipose tissue and atherosclerosis. Curr Vasc Pharmacol 2009; 7: 169–179, doi: 10.2174/157016109787455680.
https://doi.org/10.2174/1570161097874556...
). The available anti-obesity drugs such as orlistat and sibutramine (SIB) have modest clinical efficacy, but safety and tolerability concerns may limit their use (33. Ling H, Lenz TL, Burns TL, Hilleman DE. Reducing the risk of obesity: defining the role of weight loss drugs. Pharmacotherapy 2013; 33: 1308–1321, doi: 10.1002/phar.1277.
https://doi.org/10.1002/phar.1277...
,44. Sumithran P, Proietto J. Benefit-risk assessment of orlistat in the treatment of obesity. Drug Saf 2014; 37: 597–608, doi: 10.1007/s40264-014-0210-7.
https://doi.org/10.1007/s40264-014-0210-...
). Therefore, there is a need for the discovery and development of novel, safe, and effective drugs for the control and treatment of obesity.

Due to the increasing consumer knowledge of the link between diet and health, there is an increased awareness and demand for functional food ingredients and nutraceuticals. This leads to a mindset directed to self-medication to avoid the undesirable side effects associated with consumption of synthesized drugs and also to avoid the increasing cost of drug therapy. Several studies have focused on prevention and treatment of obesity and its associated health risks using plant-derived phenolic compounds. The nutritional effects of phenolic compounds have been well established in their role in modulating specific physiological functions in rodents and human beings (55. Cheng DM, Pogrebnyak N, Kuhn P, Poulev A, Waterman C, Rojas-Silva P, et al. Polyphenol-rich Rutgers Scarlet Lettuce improves glucose metabolism and liver lipid accumulation in diet-induced obese C57BL/6 mice. Nutrition 2014; 30: S52-S58, doi: 10.1016/j.nut.2014.02.022.
https://doi.org/10.1016/j.nut.2014.02.02...
,66. Alam MA, Subhan N, Hossain H, Hossain M, Reza HM, Rahman MM, et al. Hydroxycinnamic acid derivatives: a potential class of natural compounds for the management of lipid metabolism and obesity. Nutr Metab 2016; 13: 27, doi: 10.1186/s12986-016-0080-3.
https://doi.org/10.1186/s12986-016-0080-...
). Ferulic acid (FA) (Figure 1) belongs to the family of phenolic acids and is very abundant in fruits, vegetables and grains, such as rice bran, oats, wheat, barley, roasted coffee, tomatoes, asparagus, berries, vegetables, citrus fruits and leaves of most plants. FA has shown to exhibit hypocholesterolemic, hypoglycemic, anti-atherogenic, anticancer, antioxidant, antidiabetic and anti-inflammatory properties in experimental studies (77. Zhao Z, Moghadasian MH. Chemistry, natural sources, dietary intake and pharmacokinetic properties of ferulic acid: A review. Food Chem 2008; 109: 691–702, doi: 10.1016/j.foodchem.2008.02.039.
https://doi.org/10.1016/j.foodchem.2008....
1212. Ramar M, Manikandan B, Raman T, Priyadarsini A, Palanisamy S, Velayudam M, et al. Protective effect of ferulic acid and resveratrol against alloxan-induced diabetes in mice. Eur J Pharmacol 2012; 690: 226–235, doi: 10.1016/j.ejphar.2012.05.019.
https://doi.org/10.1016/j.ejphar.2012.05...
). Moreover, FA is a phenolic acid of low toxicity and it can be absorbed, easily metabolized in the human body (1313. Zhao Z, Egashira Y, Sanada H. Ferulic acid is quickly absorbed from rat stomach as the free form and then conjugated mainly in liver. J Nutr 2004; 134: 3083–3088.). Recently, FA has also shown to improve the glucose and lipid homeostasis in high-fat diet (HFD)-fed mice probably via modulating the expression of lipogenic and gluconeogenic genes in liver tissues (1414. Naowaboot J, Piyabhan P, Munkong N, Parklak W, Pannangpetch P. Ferulic acid improves lipid and glucose homeostasis in high-fat diet-induced obese mice. Clin Exp Pharmacol Physiol 2016; 43: 242–250, doi: 10.1111/1440-1681.12514.
https://doi.org/10.1111/1440-1681.12514...
).

Figure 1
Chemical structure of ferulic acid ((E)-3-(4-hydroxy-3-methoxy-phenyl)prop-2-enoic acid).

The consumption of fat-rich foods may activate an inflammatory response in the hypothalamus, thereby disturbing the anorexigenic and thermogenic signals generated by the hormones, ghrelin, leptin and insulin, leading to anomalous body mass control (1515. Velloso LA, Araujo EP, de Souza CT. Diet-induced inflammation of the hypothalamus in obesity. Neuroimmunomodulation 2008; 15: 189–193, doi: 10.1159/000153423.
https://doi.org/10.1159/000153423...
). Since FA exhibits anti-oxidant (99. Srinivasan M, Sudheer AR, Menon VP. Ferulic acid: therapeutic potential through its antioxidant property. J Clin Biochem Nutr 2007; 40: 92–100, doi: 10.3164/jcbn.40.92.
https://doi.org/10.3164/jcbn.40.92...
10. 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: 9800–9804, doi: 10.1021/jf0714463.
https://doi.org/10.1021/jf0714463...
11. Jin Son M, W Rico C, 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: 150–156, doi: 10.3164/jcbn.09-98.
https://doi.org/10.3164/jcbn.09-98...
) and anti-inflammatory properties (1616. Das U, Manna K, Sinha M, Datta S, Das DK, Chakraborty A, et al. Role of ferulic acid in the amelioration of ionizing radiation induced inflammation: a murine model. PLoS One 2014; 9: e97599, doi: 10.1371/journal.pone.0097599.
https://doi.org/10.1371/journal.pone.009...
), and modulatory effects on glucose and lipid metabolism (1414. Naowaboot J, Piyabhan P, Munkong N, Parklak W, Pannangpetch P. Ferulic acid improves lipid and glucose homeostasis in high-fat diet-induced obese mice. Clin Exp Pharmacol Physiol 2016; 43: 242–250, doi: 10.1111/1440-1681.12514.
https://doi.org/10.1111/1440-1681.12514...
,1717. Mancuso C, Santangelo R. Ferulic acid: pharmacological and toxicological aspects. Food Chem Toxicol 2014; 65: 185–195, doi: 10.1016/j.fct.2013.12.024.
https://doi.org/10.1016/j.fct.2013.12.02...
), it is likely that FA modulates adipogenesis. Therefore, the present study was aimed to demonstrate the anti-obesity effect of FA (10 mg/kg) compared to sibutramine (10 mg/kg), a well-known weight loss agent, using the mouse model of HFD-induced obesity.

Material and Methods

Chemicals and drugs

Ferulic acid was purchased from Sigma Aldrich® (USA), 98% pure. Sibutramine was purchased from Aché® (Brazil). All other chemicals and reagents used were of analytical grade and obtained from standard commercial suppliers.

Animals

Male Swiss mice (32 total, weighing 20–25 g) obtained from the Central Animal House of Universidade Federal do Ceará were used. They were kept in propylene cages, at a room temperature (24±2°C) on a 12-h light/dark cycle with food (chow) and water provided ad libitum. Experimental protocols (#34/2011) were approved by the Universidade Federal do Ceará Institutional Committee on Care and Use of Animals for Experimentation, in accordance with the guidelines of the National Institutes of Health, USA.

Diet composition

The normal diet (ND) used was the pelleted chow obtained from a commercial source (Nuvilab, Brazil). It consisted of 19.0% protein, 56% carbohydrate, 3.5% lipids, 4.5% cellulose, 5.0% vitamins and minerals, and 12% humidity with a total energy content of 17.03 kJ/g. The standardized HFD used for the study (1818. Estadella D, Oyama LM, Damaso AR, Ribeiro EB, Oller Do Nascimento CM. Effect of palatable hyperlipidic diet on lipid metabolism of sedentary and exercised rats. Nutrition 2004; 20: 218–224, doi: 10.1016/j.nut.2003.10.008.
https://doi.org/10.1016/j.nut.2003.10.00...
) comprised the following hypercaloric constituents: 15 g of laboratory animal chow, 10 g of roasted ground nut, 10 g of milk chocolate, and 5 g of maizen cookies. These ingredients were ground and prepared in the form of pellets that contained, by weight, 20% protein, 48% carbohydrate, 20% lipids, 4% cellulose, 5% vitamins and minerals and 3% humidity. The net energy content of this diet was 21.40 kJ/g. Thus, the HFD, compared with ND, was hypercaloric, and it contained less carbohydrate but more lipids with a net energy difference of 4.37 kJ/g. To avoid auto-oxidation of the fat components, food was stored at approximately 24°C.

Anti-obesity activity

Mice were randomly divided into four groups (n=8) matched for body weight after 1 week being fed laboratory pellet chow. The control group (ND) continued to be fed laboratory pellet chow ad libitum. The remaining mice consumed HFD (HFD control), HFD+FA (0.05% in drinking water, which is equivalent to 10 mg/kg of body weight based on water consumption), or HFD+SIB (0.05% in drinking water, which amounts to 10 mg/kg of body weight) for 15 weeks. FA and SIB concentrations were based on our previous experiments (88. 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: 2825–2830, doi: 10.1021/jf072896y.
https://doi.org/10.1021/jf072896y...
,1919. Carvalho KM, Marinho Filho JD, de Melo TS, Araujo AJ, Quetz JS, da Cunha MP, et al. The resin from protium heptaphyllum prevents high-fat diet-induced obesity in mice: scientific evidence and potential mechanisms. Evid Based Complement Alternat Med 2015; 2015: 106157, doi: 10.1155/2015/106157.
https://doi.org/10.1155/2015/106157...
). FA was suspended initially in 3% (v/v) Tween 80 and then further diluted in water. HFD-fed controls received the same vehicle. As SIB is water-soluble, no vehicle was used. FA- or vehicle-containing water was changed twice a week, and weekly consumption of water (mL/week) was recorded. The body weight of each mouse was measured once a week, the total amount of food consumption was recorded every day for 15 weeks, and weekly consumption of food (g/week) was calculated. At the end of this period, animals were fast for 6 h, blood was taken by venous puncture and then they were killed by cervical dislocation. The plasma was prepared and either used within a few hours or frozen at -70°C until analysis. The liver and visceral adipose tissues (epididymal and parametrial) were dissected, weighed, and expressed in mg/10 g of body weight.

Biochemical analysis

Plasma amylase and lipase activities were determined by a kinetic method using commercial kits for amylase (Labtest®, Brazil) and lipase (Bioclin®, Brazil). The assays were performed according to the manufacturer’s instructions, and their levels are reported in U/L. Plasma glucose, triglycerides, and total cholesterol were analyzed using commercial kits (Labtest®), and the levels are reported as mg/dL. Plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST), reported in U/L, were analyzed by a kinetic method using commercial kits (Labtest®). Plasma TNF-α, MCP-1, insulin, leptin, and ghrelin levels were measured by enzyme linked immunosorbent assays (Crystal Chem, USA) performed in duplicate and reported in ng/mL or pg/mL.

Non-protein sulfhydryls (NP-SH)

NP-SH (non-protein sulfhydrils, GSH) in hepatic tissues were determined by Ellman’s reaction using 5’5’-dithio-bis-2-nitrobenzoic acid (DTNB) (2020. Sedlak J, Lindsay RH. Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman's reagent. Anal Biochem 1968; 25: 192–205, doi: 10.1016/0003-2697(68)90092-4.
https://doi.org/10.1016/0003-2697(68)900...
). Aliquots of 4 mL of the homogenates in ice-cold ethylenediaminetetraacetic acid (EDTA; 0.02 mol/L, pH 8.9) were mixed with 3.2 mL of distilled water and 0.8 mL of 50% trichloroacetic acid (TCA). The tubes were centrifuged at 800 g for 15 min at 4°C. The supernatant (2 mL) was mixed with 4 mL Tris buffer (0.4 mol/L, pH 8.9) and 0.1 mL of DTNB (0.01 mol/L). The absorbance was measured within 5 min after addition of DTNB at 412 nm. The absorbance values were extrapolated from a glutathione standard curve and reported as µg/g of hepatic tissue.

Malondialdehyde assay

The concentration of hepatic lipid peroxidation was determined by estimating malondialdehyde (MDA) using the thiobarbituric acid test (2121. Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 1979; 95: 351–358, doi: 10.1016/0003-2697(79)90738-3.
https://doi.org/10.1016/0003-2697(79)907...
). The hepatic tissue was homogenized in 0.15 KCl, pH 7.4. The homogenate was maintained in a water bath for 60 min at 37°C. Perchloric acid (35%) was added to the homogenate and centrifuged at 17,500 g for 10 min at 4°C. The supernatant was mixed with 1.2% thiobarbituric acid, and the mixture was heated at 98°C for 30 min. After cooling to room temperature, the absorbance was measured at 532 nm. The standard curve was obtained using 1,1,3,3-tetramethoxypropane. The results were reported as nmol of MDA/g hepatic tissue.

Histological analysis

The epididymal fat pads were excised, fixed in 10% formalin, and processed routinely for paraffin embedding. Tissue sections of 5-µm thick were cut, processed for hematoxylin and eosin (H&E) staining, and examined under a light microscope for histological changes.

Statistical analyses

The results are reported as means±SE for 8 animals in each group. Data were analyzed by one-way analysis of variance (ANOVA) followed by Student-Newman-Keuls test, using the GraphPad (USA) Prism program (version 5.0). Differences were considered to be significant at P<0.05.

Results

Antiobesity effect of ferulic acid and sibutramine in mice fed HFD

The body weight of mice in the four groups was similar at baseline and constantly increased over time. As shown in Figure 2A, at the end of the 15-week experimental period, the high calorie diet induced a significant (P<0.05) increase of 29% in body weight of the HFD group compared to the ND group (54.88±2 vs 42.36±1.27 g, respectively). Abdominal fat weight, which includes the epididymal and parametrial pads, was significantly (P<0.05) higher in HFD controls compared to ND controls (Figure 2B). FA supplementation in drinking water (50 mg/L) significantly (P<0.05) reduced the final body weight (g) and abdominal fat deposition (mg/10 g) to 42.56±1.54 and 31.95±3.73, respectively, when compared to the HFD group, representing reductions of 22 and 38% in these parameters. The FA had an effect similar to sibutramine in reducing body weight and abdominal fat accumulation (23 and 36%, respectively). While treatment with FA and SIB slightly but significantly (P<0.05) reduced water consumption compared to ND and HFD controls, both treatments effectively decreased the net energy intake almost to the level seen in ND fed mice when compared to values observed in HFD controls (Table 1).

Figure 2
Effect of ferulic acid (FA) and sibutramine (SIB) on body weight (A) and abdominal fat content (B) in mice fed a HFD during 15 weeks. Results are reported as means±SE. ND: normal diet; HFD: high-fat diet. a, P<0.05 vs ND; b, P<0.05 vs HFD (one-way ANOVA followed by Student-Newman-Keuls test).

Effects of ferulic acid and sibutramine on plasma parameters

As shown in Table 2, plasma levels of glucose and insulin were significantly (P<0.05) higher (78% and 3.2-fold, respectively) in the HFD group compared to ND group. FA treatment for 15 weeks led to a significant (P<0.05) 33% reduction in plasma glucose levels and 58% reduction in plasma insulin levels compared to HFD group. Treatment with SIB showed no significant influence on plasma glucose, but it effectively decreased insulin level (48%). Compared to the ND group, the HFD group significantly (P<0.05) decreased plasma ghrelin (orexigenic hormone), while it elevated the leptin (anorexigenic hormone) level (Table 1). These changes induced by the HFD were effectively reversed by FA treatment. SIB treatment, however, caused no significant change in the level of ghrelin hormone.

The amylase and lipase activities were significantly (P<0.05) higher in the HFD group (65 and 37%, respectively), compared to ND group. FA treatment caused significant (P<0.05) reductions in the activities of both amylase and lipase by 42 and 27%, respectively. SIB, used as a reference standard, reduced significantly (P<0.05) the amylase activity (38%), while it had no influence on lipase activity (Table 2).

The HFD raised total cholesterol (70%) and triglycerides (66%) significantly (P<0.05) compared with the ND. These increased levels of the HFD group were significantly (P<0.05) lowered by FA and SIB treatments (Table 2).

Effects of ferulic acid and sibutramine on plasma levels of inflammation and lipid peroxidation-related parameters

Table 3 shows the effects of FA and SIB treatments on plasma levels of monocyte chemotactic protein 1 (MCP-1) and tumor necrosis factor (TNF-α), and hepatic levels of NP-SH, and MDA, in mice fed experimental diets for 15 weeks. The levels of MCP-1 showed an increase of 55% in the HFD relative to ND group, which was significantly (P<0.05) decreased in groups treated with FA and SIB (44 and 34%, respectively). Similarly, the increase in TNF-α promoted by the HFD was also greatly decreased in groups treated with FA and SIB. The hepatic tissue levels of NP-SH, and MDA were significantly (P<0.05) elevated by the HFD, whereas in mice treated with FA or SIB these changes were partially reversed, but not significantly.

Effects of ferulic acid and sibutramine on satiety parameters

There were no significant changes in serum urea and creatinine levels among all tested groups. However, animals subjected to the hypercaloric diet showed an increase in the activity of ALT and AST (41 and 46%, respectively), indicating a possible liver damage (Table 4). Serum ALT and AST decreased significantly (P<0.05) in groups treated with FA compared to HFD control group, representing a decrease of 33 and 74% in the activity of the respective enzymes. Sibutramine, used as positive control, also significantly (P<0.05) reduced the activity of ALT and AST by 36 to 59% compared to HFD fed animals.

Effects of ferulic acid and sibutramine on epididymal adipocytes size in mice fed HFD

Mice fed HFD for 15 weeks apparently presented greater number of epididymal fat cells with an increased size of adipocyte (Figure 3) compared to the group that received normal diet. In the groups treated with FA or SIB, the adipocyte size almost resembled those of the ND-fed mice.

Figure 3
Histology of adipose tissue of mice fed the experimental diets for 15 weeks. Representative microphotographs of mouse epididymal fat pad (A), normal diet showing normal architecture of adipocytes (B), high-fat diet showing an increased size adipocyte (C), high-fat diet+ferulic acid treatment, and (D), high-fat diet+sibutramine treatment, which presents smaller adipocytes compared to normal diet-fed animals (H&E, ×100).

Discussion

Past studies have addressed the hypoglycemic, hypolipidemic, and antioxidant properties of FA in HFD fed animals (88. 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: 2825–2830, doi: 10.1021/jf072896y.
https://doi.org/10.1021/jf072896y...
,99. Srinivasan M, Sudheer AR, Menon VP. Ferulic acid: therapeutic potential through its antioxidant property. J Clin Biochem Nutr 2007; 40: 92–100, doi: 10.3164/jcbn.40.92.
https://doi.org/10.3164/jcbn.40.92...
), but its influence in lowering the visceral adiposity has not been analyzed and reported. The results obtained in this study clearly showed that a HFD for 15 weeks promoted visceral adiposity and weight gain in Swiss mice, and treatments with the FA, or a known anorectic agent, such as SIB (2222. Araujo JR, Martel F. Sibutramine effects on central mechanisms regulating energy homeostasis. Curr Neuropharmacol 2012; 10: 49–52, doi: 10.2174/157015912799362788.
https://doi.org/10.2174/1570159127993627...
) in drinking water (50 mg/L) prevented this adiposity and weight gain.

Ghrelin from the stomach and leptin from adipose tissue regulate appetite and energy homeostasis in humans and rodents. It has been observed that the long-term intake of a HFD can induce hyperleptinemia and hypoghrelinemia and a significant positive correlation between plasma leptin levels and epididymal fat mass (2323. Handjieva-Darlenska T, Boyadjieva N. The effect of high-fat diet on plasma ghrelin and leptin levels in rats. J Physiol Biochem 2009; 65: 157–164, doi: 10.1007/BF03179066.
https://doi.org/10.1007/BF03179066...
). Consistent with these earlier findings, the present study with mice on HFD for 15 weeks demonstrated hyperleptinemia and hypoghrelinemia and an increased abdominal fat deposition, which could be counteracted by FA supplementation. The hypothalamus is considered the major site of anorexigenic and orexigenic signaling integration through activation of respective receptors for ghrelin (orexigenic) and leptin (anorexigenic). Obesity is associated with elevated leptin and resistance to leptin effects on energy homeostasis (2424. Pan H, Guo J, Su Z. Advances in understanding the interrelations between leptin resistance and obesity. Physiol Behav 2014; 130: 157–169, doi: 10.1016/j.physbeh.2014.04.003.
https://doi.org/10.1016/j.physbeh.2014.0...
). Our current study demonstrated that FA supplementation significantly lowers circulating leptin level. Since FA supplementation could reduce the food and energy intakes, we assume that the HFD-induced changes in ghrelin and leptin plasma levels may, in part, account for the observed body weight loss and reduced abdominal fat. However, the underlying molecular mechanism of FA in its antiobese effect remains to be established.

Several studies demonstrated that adipose tissue dysregulation and aberrant adipokine secretion contribute to low-grade chronic proinflammatory state and insulin resistance (2525. Samaras K, Botelho NK, Chisholm DJ, Lord RV. Subcutaneous and visceral adipose tissue gene expression of serum adipokines that predict type 2 diabetes. Obesity 2010; 18: 884–889, doi: 10.1038/oby.2009.443.
https://doi.org/10.1038/oby.2009.443...
26. Bremer AA, Jialal I. Adipose tissue dysfunction in nascent metabolic syndrome. J Obes 2013; 2013: 393192, doi: 10.1155/2013/393192.
https://doi.org/10.1155/2013/393192...
2727. Fisman EZ, Tenenbaum A. Adiponectin: a manifold therapeutic target for metabolic syndrome, diabetes, and coronary disease? Cardiovasc Diabetol 2014; 13: 103, doi: 10.1186/1475-2840-13-103.
https://doi.org/10.1186/1475-2840-13-103...
). Obesity has been considered a disease in which there is a predominance of proinflammatory cytokines. Recent findings suggest that the size of adipocytes is a major modulator of their endocrine function. Hypertrophic adipocytes secrete greater amounts of TNF-α and MCH-1 than normal adipocytes, and this excess secretion has been hypothesized to cause insulin resistance (2828. Liang H, Yin B, Zhang H, Zhang S, Zeng Q, Wang J, et al. Blockade of tumor necrosis factor (TNF) receptor type 1-mediated TNF-alpha signaling protected Wistar rats from diet-induced obesity and insulin resistance. Endocrinology 2008; 149: 2943–2951, doi: 10.1210/en.2007-0978.
https://doi.org/10.1210/en.2007-0978...
29. Baranowski M, Enns J, Blewett H, Yakandawala U, Zahradka P, Taylor CG. Dietary flaxseed oil reduces adipocyte size, adipose monocyte chemoattractant protein-1 levels and T-cell infiltration in obese, insulin-resistant rats. Cytokine 2012; 59: 382–391, doi: 10.1016/j.cyto.2012.04.004.
https://doi.org/10.1016/j.cyto.2012.04.0...
3030. Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006; 17: 4–12.). In our studies, circulating levels of MCP-1 and TNF-α, as well as insulin were greatly elevated in mice fed on HFD for 15 weeks, indicating a proinflammatory state and insulin resistance seen in type-2 diabetes. Both FA and SIB reduced the adipocyte size, as well as the circulating levels of adipokines, TNF-α and MCH-1, which would explain its ameliorating effect on abdominal adiposity and insulin resistance.

In addition, there was a significant improvement in biochemical parameters such as plasma glucose, cholesterol and triglycerides, as well as the lipase and amylase activities, when mice were treated with the combination of FA plus HFD. Inhibition of pancreatic lipase and the associated reduction of lipid absorption is an attractive approach for the discovery of potent agents (3131. Buchholz T, Melzig MF. Polyphenolic compounds as pancreatic lipase inhibitors. Planta Med 2015; 81: 771–783, doi: 10.1055/s-0035-1565716.
https://doi.org/10.1055/s-0035-1565716...
). Currently, the only clinically approved pharmacological agent for pancreatic lipase inhibitor is orlistat. However, its usage is compromised by unpleasant gastrointestinal adverse reactions (oily stools and flatulence). An important target for the treatment of obesity includes the development of inhibitors of nutrient digestion and absorption. FA is therefore capable of modulating glucose and lipid metabolism, consistent with earlier reports (88. 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: 2825–2830, doi: 10.1021/jf072896y.
https://doi.org/10.1021/jf072896y...
,1616. Das U, Manna K, Sinha M, Datta S, Das DK, Chakraborty A, et al. Role of ferulic acid in the amelioration of ionizing radiation induced inflammation: a murine model. PLoS One 2014; 9: e97599, doi: 10.1371/journal.pone.0097599.
https://doi.org/10.1371/journal.pone.009...
,3232. Kesh SB, Sikder K, Manna K, Das DK, Khan A, Das N, et al. Promising role of ferulic acid, atorvastatin and their combination in ameliorating high fat diet-induced stress in mice. Life Sci 2013; 92: 938–949, doi: 10.1016/j.lfs.2013.03.015.
https://doi.org/10.1016/j.lfs.2013.03.01...
).

Overweight and obesity have a major impact on global health. Strategies for weight control management affect gut hormones as potential targets for the appetite metabolic regulation and stimulation of energy expenditure. Past and current weight-loss medications have serious safety risks. For example, orlistat has a good safety profile but a high rate of gastrointestinal side effects (44. Sumithran P, Proietto J. Benefit-risk assessment of orlistat in the treatment of obesity. Drug Saf 2014; 37: 597–608, doi: 10.1007/s40264-014-0210-7.
https://doi.org/10.1007/s40264-014-0210-...
). Another example is Garcinia Cambogia that although it protects against HFD-induced obesity by modulating adipose fatty acid synthesis and β-oxidation, it can induce hepatic fibrosis, inflammation and oxidative stress (3333. Kim YJ, Choi MS, Park YB, Kim SR, Lee MK, Jung UJ. Garcinia Cambogia attenuates diet-induced adiposity but exacerbates hepatic collagen accumulation and inflammation. World J Gastroenterol 2013; 19: 4689–4701.). In this study, the antiobesity potential of FA is almost comparable to SIB, a weight loss promoting agent that was banned in several countries due to cardiovascular toxicity. However, studies are still relevant as it is often a hidden ingredient in herbal and over the counter slimming products (3434. Oberholzer HM, Van Der Schoor C, Bester MJ. Sibutramine, a serotonin-norepinephrine reuptake inhibitor, causes fibrosis in rats. Environ Toxicol Pharmacol 2015; 40: 71–76. doi: 10.1016/j.etap.2015.05.011.
https://doi.org/10.1016/j.etap.2015.05.0...
). In this context, there are reports that FA may have health benefits by minimizing the cardiovascular complications of metabolic syndrome (3535. Senaphan K, Kukongviriyapan U, Sangartit W, Pakdeechote P, Pannangpetch P, Prachaney P, et al. Ferulic acid alleviates changes in a rat model of metabolic syndrome induced by high-carbohydrate, high-fat diet. Nutrients 2015; 7: 6446–6464, doi: 10.3390/nu7085283.
https://doi.org/10.3390/nu7085283...
). Besides the antiobese potential of FA, this study also evaluated its likely toxicity to liver and kidney. Elevated serum ALT and AST activities are biomarkers of liver injury (3636. Kamei T, Asano K, Nakamura S. Determination of serum glutamate oxaloacetate transaminase and glutamate pyruvate transaminase by using L-glutamate oxidase. Chem Pharm Bull 1986; 34: 409–412, doi: 10.1248/cpb.34.409.
https://doi.org/10.1248/cpb.34.409...
). ALT and AST were both found to be raised significantly in mice fed the HFD alone compared with ND controls, while FA inhibited this HFD-induced increase. Further, FA did not alter circulating rates of urea and creatinine, which confirms no potential kidney or hepatic damage. Moreover, it prevented HFD-induced fatty liver in the present study and offered protection against diosbulbin-induced hepatotoxicity (3737. Wang JM, Sheng YC, Ji LL, Wang ZT. Ferulic acid prevents liver injury and increases the anti-tumor effect of diosbulbin B in vivo. J Zhejiang Univ Sci B 2014; 15: 540–547, doi: 10.1631/jzus.B1300250.
https://doi.org/10.1631/jzus.B1300250...
) and glycerol-induced nephrotoxicity (3838. Manikandan R, Beulaja M, Thiagarajan R, Pandi M, Arulvasu C, Prabhu NM, et al. Ameliorative effect of ferulic acid against renal injuries mediated by nuclear factor-kappaB during glycerol-induced nephrotoxicity in Wistar rats. Ren Fail 2014; 36: 154–165, doi: 10.3109/0886022X.2013.835223.
https://doi.org/10.3109/0886022X.2013.83...
) in other studies.

This study has its strength in the evaluation of various parameters that are closely related to metabolic syndrome and obesity, which were improved with FA treatment. Adipose tissue and liver are the two main organs involved in lipid metabolism, and transcriptional control of gene expression is a common mechanism by which lipids and other nutrients affect metabolism. The limitation of this study is the absence of analysis of possible genetic changes related to obesity promoted by FA treatment. Therefore, further studies are required to elucidate the antiobesity FA action.

In summary, ferulic acid effectively prevented high fat diet-induced visceral adiposity and body weight gain via mechanisms involving the modulation of food regulatory peptide hormones (insulin, ghrelin and leptin), inhibition of serum amylase and lipase activity, and suppression of adipocyte-derived pro-inflammatory cytokines MCP-1 and TNF-α. These effects of ferulic acid may be beneficial and therefore it might be a promising adjuvant therapy for the treatment of obesity and its complications.

Acknowledgments

The authors would like to thank Aguinéa Rocha de Morais for technical support. This research was supported by the grants and fellowships from CNPq and FUNCAP.

References

  • 1
    Caveney E, Caveney BJ, Somaratne R, Turner JR, Gourgiotis L. Pharmaceutical interventions for obesity: a public health perspective. Diabetes Obes Metab 2011; 13: 490–497, doi: 10.1111/j.1463-1326.2010.01353.x.
    » https://doi.org/10.1111/j.1463-1326.2010.01353.x
  • 2
    Ohman MK, Wright AP, Wickenheiser KJ, Luo W, Eitzman DT. Visceral adipose tissue and atherosclerosis. Curr Vasc Pharmacol 2009; 7: 169–179, doi: 10.2174/157016109787455680.
    » https://doi.org/10.2174/157016109787455680
  • 3
    Ling H, Lenz TL, Burns TL, Hilleman DE. Reducing the risk of obesity: defining the role of weight loss drugs. Pharmacotherapy 2013; 33: 1308–1321, doi: 10.1002/phar.1277.
    » https://doi.org/10.1002/phar.1277
  • 4
    Sumithran P, Proietto J. Benefit-risk assessment of orlistat in the treatment of obesity. Drug Saf 2014; 37: 597–608, doi: 10.1007/s40264-014-0210-7.
    » https://doi.org/10.1007/s40264-014-0210-7
  • 5
    Cheng DM, Pogrebnyak N, Kuhn P, Poulev A, Waterman C, Rojas-Silva P, et al. Polyphenol-rich Rutgers Scarlet Lettuce improves glucose metabolism and liver lipid accumulation in diet-induced obese C57BL/6 mice. Nutrition 2014; 30: S52-S58, doi: 10.1016/j.nut.2014.02.022.
    » https://doi.org/10.1016/j.nut.2014.02.022
  • 6
    Alam MA, Subhan N, Hossain H, Hossain M, Reza HM, Rahman MM, et al. Hydroxycinnamic acid derivatives: a potential class of natural compounds for the management of lipid metabolism and obesity. Nutr Metab 2016; 13: 27, doi: 10.1186/s12986-016-0080-3.
    » https://doi.org/10.1186/s12986-016-0080-3
  • 7
    Zhao Z, Moghadasian MH. Chemistry, natural sources, dietary intake and pharmacokinetic properties of ferulic acid: A review. Food Chem 2008; 109: 691–702, doi: 10.1016/j.foodchem.2008.02.039.
    » https://doi.org/10.1016/j.foodchem.2008.02.039
  • 8
    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: 2825–2830, doi: 10.1021/jf072896y.
    » https://doi.org/10.1021/jf072896y
  • 9
    Srinivasan M, Sudheer AR, Menon VP. Ferulic acid: therapeutic potential through its antioxidant property. J Clin Biochem Nutr 2007; 40: 92–100, doi: 10.3164/jcbn.40.92.
    » https://doi.org/10.3164/jcbn.40.92
  • 10
    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: 9800–9804, doi: 10.1021/jf0714463.
    » https://doi.org/10.1021/jf0714463
  • 11
    Jin Son M, W Rico C, 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: 150–156, doi: 10.3164/jcbn.09-98.
    » https://doi.org/10.3164/jcbn.09-98
  • 12
    Ramar M, Manikandan B, Raman T, Priyadarsini A, Palanisamy S, Velayudam M, et al. Protective effect of ferulic acid and resveratrol against alloxan-induced diabetes in mice. Eur J Pharmacol 2012; 690: 226–235, doi: 10.1016/j.ejphar.2012.05.019.
    » https://doi.org/10.1016/j.ejphar.2012.05.019
  • 13
    Zhao Z, Egashira Y, Sanada H. Ferulic acid is quickly absorbed from rat stomach as the free form and then conjugated mainly in liver. J Nutr 2004; 134: 3083–3088.
  • 14
    Naowaboot J, Piyabhan P, Munkong N, Parklak W, Pannangpetch P. Ferulic acid improves lipid and glucose homeostasis in high-fat diet-induced obese mice. Clin Exp Pharmacol Physiol 2016; 43: 242–250, doi: 10.1111/1440-1681.12514.
    » https://doi.org/10.1111/1440-1681.12514
  • 15
    Velloso LA, Araujo EP, de Souza CT. Diet-induced inflammation of the hypothalamus in obesity. Neuroimmunomodulation 2008; 15: 189–193, doi: 10.1159/000153423.
    » https://doi.org/10.1159/000153423
  • 16
    Das U, Manna K, Sinha M, Datta S, Das DK, Chakraborty A, et al. Role of ferulic acid in the amelioration of ionizing radiation induced inflammation: a murine model. PLoS One 2014; 9: e97599, doi: 10.1371/journal.pone.0097599.
    » https://doi.org/10.1371/journal.pone.0097599
  • 17
    Mancuso C, Santangelo R. Ferulic acid: pharmacological and toxicological aspects. Food Chem Toxicol 2014; 65: 185–195, doi: 10.1016/j.fct.2013.12.024.
    » https://doi.org/10.1016/j.fct.2013.12.024
  • 18
    Estadella D, Oyama LM, Damaso AR, Ribeiro EB, Oller Do Nascimento CM. Effect of palatable hyperlipidic diet on lipid metabolism of sedentary and exercised rats. Nutrition 2004; 20: 218–224, doi: 10.1016/j.nut.2003.10.008.
    » https://doi.org/10.1016/j.nut.2003.10.008
  • 19
    Carvalho KM, Marinho Filho JD, de Melo TS, Araujo AJ, Quetz JS, da Cunha MP, et al. The resin from protium heptaphyllum prevents high-fat diet-induced obesity in mice: scientific evidence and potential mechanisms. Evid Based Complement Alternat Med 2015; 2015: 106157, doi: 10.1155/2015/106157.
    » https://doi.org/10.1155/2015/106157
  • 20
    Sedlak J, Lindsay RH. Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman's reagent. Anal Biochem 1968; 25: 192–205, doi: 10.1016/0003-2697(68)90092-4.
    » https://doi.org/10.1016/0003-2697(68)90092-4
  • 21
    Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 1979; 95: 351–358, doi: 10.1016/0003-2697(79)90738-3.
    » https://doi.org/10.1016/0003-2697(79)90738-3
  • 22
    Araujo JR, Martel F. Sibutramine effects on central mechanisms regulating energy homeostasis. Curr Neuropharmacol 2012; 10: 49–52, doi: 10.2174/157015912799362788.
    » https://doi.org/10.2174/157015912799362788
  • 23
    Handjieva-Darlenska T, Boyadjieva N. The effect of high-fat diet on plasma ghrelin and leptin levels in rats. J Physiol Biochem 2009; 65: 157–164, doi: 10.1007/BF03179066.
    » https://doi.org/10.1007/BF03179066
  • 24
    Pan H, Guo J, Su Z. Advances in understanding the interrelations between leptin resistance and obesity. Physiol Behav 2014; 130: 157–169, doi: 10.1016/j.physbeh.2014.04.003.
    » https://doi.org/10.1016/j.physbeh.2014.04.003
  • 25
    Samaras K, Botelho NK, Chisholm DJ, Lord RV. Subcutaneous and visceral adipose tissue gene expression of serum adipokines that predict type 2 diabetes. Obesity 2010; 18: 884–889, doi: 10.1038/oby.2009.443.
    » https://doi.org/10.1038/oby.2009.443
  • 26
    Bremer AA, Jialal I. Adipose tissue dysfunction in nascent metabolic syndrome. J Obes 2013; 2013: 393192, doi: 10.1155/2013/393192.
    » https://doi.org/10.1155/2013/393192
  • 27
    Fisman EZ, Tenenbaum A. Adiponectin: a manifold therapeutic target for metabolic syndrome, diabetes, and coronary disease? Cardiovasc Diabetol 2014; 13: 103, doi: 10.1186/1475-2840-13-103.
    » https://doi.org/10.1186/1475-2840-13-103
  • 28
    Liang H, Yin B, Zhang H, Zhang S, Zeng Q, Wang J, et al. Blockade of tumor necrosis factor (TNF) receptor type 1-mediated TNF-alpha signaling protected Wistar rats from diet-induced obesity and insulin resistance. Endocrinology 2008; 149: 2943–2951, doi: 10.1210/en.2007-0978.
    » https://doi.org/10.1210/en.2007-0978
  • 29
    Baranowski M, Enns J, Blewett H, Yakandawala U, Zahradka P, Taylor CG. Dietary flaxseed oil reduces adipocyte size, adipose monocyte chemoattractant protein-1 levels and T-cell infiltration in obese, insulin-resistant rats. Cytokine 2012; 59: 382–391, doi: 10.1016/j.cyto.2012.04.004.
    » https://doi.org/10.1016/j.cyto.2012.04.004
  • 30
    Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H, et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 2006; 17: 4–12.
  • 31
    Buchholz T, Melzig MF. Polyphenolic compounds as pancreatic lipase inhibitors. Planta Med 2015; 81: 771–783, doi: 10.1055/s-0035-1565716.
    » https://doi.org/10.1055/s-0035-1565716
  • 32
    Kesh SB, Sikder K, Manna K, Das DK, Khan A, Das N, et al. Promising role of ferulic acid, atorvastatin and their combination in ameliorating high fat diet-induced stress in mice. Life Sci 2013; 92: 938–949, doi: 10.1016/j.lfs.2013.03.015.
    » https://doi.org/10.1016/j.lfs.2013.03.015
  • 33
    Kim YJ, Choi MS, Park YB, Kim SR, Lee MK, Jung UJ. Garcinia Cambogia attenuates diet-induced adiposity but exacerbates hepatic collagen accumulation and inflammation. World J Gastroenterol 2013; 19: 4689–4701.
  • 34
    Oberholzer HM, Van Der Schoor C, Bester MJ. Sibutramine, a serotonin-norepinephrine reuptake inhibitor, causes fibrosis in rats. Environ Toxicol Pharmacol 2015; 40: 71–76. doi: 10.1016/j.etap.2015.05.011.
    » https://doi.org/10.1016/j.etap.2015.05.011
  • 35
    Senaphan K, Kukongviriyapan U, Sangartit W, Pakdeechote P, Pannangpetch P, Prachaney P, et al. Ferulic acid alleviates changes in a rat model of metabolic syndrome induced by high-carbohydrate, high-fat diet. Nutrients 2015; 7: 6446–6464, doi: 10.3390/nu7085283.
    » https://doi.org/10.3390/nu7085283
  • 36
    Kamei T, Asano K, Nakamura S. Determination of serum glutamate oxaloacetate transaminase and glutamate pyruvate transaminase by using L-glutamate oxidase. Chem Pharm Bull 1986; 34: 409–412, doi: 10.1248/cpb.34.409.
    » https://doi.org/10.1248/cpb.34.409
  • 37
    Wang JM, Sheng YC, Ji LL, Wang ZT. Ferulic acid prevents liver injury and increases the anti-tumor effect of diosbulbin B in vivo J Zhejiang Univ Sci B 2014; 15: 540–547, doi: 10.1631/jzus.B1300250.
    » https://doi.org/10.1631/jzus.B1300250
  • 38
    Manikandan R, Beulaja M, Thiagarajan R, Pandi M, Arulvasu C, Prabhu NM, et al. Ameliorative effect of ferulic acid against renal injuries mediated by nuclear factor-kappaB during glycerol-induced nephrotoxicity in Wistar rats. Ren Fail 2014; 36: 154–165, doi: 10.3109/0886022X.2013.835223.
    » https://doi.org/10.3109/0886022X.2013.835223

Publication Dates

  • Publication in this collection
    2017

History

  • Received
    15 June 2016
  • Accepted
    4 Oct 2016
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br