ORIGINAL ARTICLE

Garcinia cambogia water extract alleviates insulin resistance and hepatic lipid accumulation in mice fed a high-fat diet

Jinya Dong1,2, Wen Li3, Xiaocui Du1,2, Xiaofang He1, Bin Deng3, Hongmei Zheng3, Yang Tian4*, Jun Sheng1,4* and Chongye Fang1,4*

1Yunnan Research Center for Advanced Tea Processing, Yunnan Agricultural University, Kunming, China; 2College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China; 3Yunnan Rural Science and Technology Service Center, Kunming, Yunnan, China; 4College of Food Science and Technology, Yunnan Agricultural University, Kunming, China

Popular scientific summary

Abstract

Background: Garcinia cambogia is widely used as a weight-loss supplement, and it is reported to be negatively associated with metabolic diseases including insulin resistance (IR), type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and dyslipidemia.

Objective: This study aimed to investigate the effect of G. cambogia water extract (GE) on high-fat diet (HFD)-induced obesity, IR, and hepatic lipid accumulation.

Design: C57BL/6 male mice were fed HFD with or without GE, GED and GEP for 16 weeks, and the mice were subjected to insulin tolerance tests and liver histological analysis. The hydroxycitric acid (HCA) levels of GE, GED, and GEP were measured by high-performance liquid chromatography.

Results: The results showed that GE significantly reduced HFD-induced body weight gain (P < 0.001), alleviated IR (P < 0.01), reduced serum total cholesterol (TC) (P < 0.001), and attenuated HFD-induced hepatic lipid accumulation. To investigate the constituent that was responsible for these effects, we separated GE into the component that dissolved in ethanol (GED) and the component that was precipitated by ethanol (GEP). Further mouse experiments showed that both GED and GEP were effective, but GED (which was used at a dose of 4 g/L) was more effective than GEP (which was used at a lower dose of 1 g/L). The HCA levels in GED and GEP were similar, although less than in GE. HCA may be the effective component in GE.

Conclusion: This study provides evidence that G. cambogia can be used as a natural supplement to alleviate IR and hepatic lipid accumulation.

Keywords: Garcinia cambogia; insulin resistance; non-alcoholic fatty liver disease; obesity; hepatic lipid accumulation; hydroxycitric acid

 

Citation: Food & Nutrition Research 2023, 67: 8977 - http://dx.doi.org/10.29219/fnr.v67.8977

Copyright: © 2023 Jinya Dong et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material for any purpose, even commercially, provided the original work is properly cited and states its license.

Received: 23 August 2022; Revised: 25 December 2022; Accepted: 4 January 2023; Published: 14 March 2023

Competing interests and funding: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. This work was supported by the Yunnan Provincial Joint Project of Basic Agricultural Research (ref. 2018FG001-036) and Yunnan International Joint Laboratory of Green Health Food (China and Thailand) (ref. 202204BI090012).

*Yang Tian, College of Food Science and Technology, Yunnan Agricultural University, No. 452, Fengyuan Road, Panlong District, Kunming 650201 China. Email: tianyang1208@163.com

*Jun Sheng, College of Food Science and Technology, Yunnan Agricultural University, No. 452, Fengyuan Road, Panlong District, Kunming 650201 China. Email: shengjun_ynau@163.com

*Chongye Fang, College of Food Science and Technology, Yunnan Agricultural University, No. 452, Fengyuan Road, Panlong District, Kunming 650201 China. Email: ffccyy2@163.com

 

Garcinia cambogia, also known as Malabar tamarind, is native to southeastern Asia (1). It is one of the most popular weight management supplements due to its beneficial effects on weight and fat reduction. Therefore, it is used in natural products for treating obesity and weight control (2, 3). Hydroxycitric acid (HCA) is a major active component of the rind of the fruit G. cambogia (4, 5). G. cambogia peel supplements contain 20–60% HCA, which may be the reason for their weight loss properties (68). HCA is an α-, β-dihydroxytricarboxylic acid that is a potent competitive inhibitor of adenosine triphosphate (ATP) citrate lyase, which is a key enzyme in the synthesis of fatty acids, cholesterol, and triglycerides (TG) (9, 10). ATP-citrate lyase cleaves citrate into oxaloacetate and acetyl-CoA, so HCA eventually limits the number of acetyl-CoA molecules available for fatty acid synthesis. HCA also decreases the accumulation of lipid droplets and the TG level by downregulating fatty acid synthase protein and increasing the phosphorylation of acetyl-CoA carboxylase (1113). HCA promotes weight loss and inhibits weight gain (without stimulating the central nervous system), stabilizes glucose levels, and improves adipokine levels (14).

Insulin resistance (IR) and non-alcoholic fatty liver disease (NAFLD) caused by high-fat diets (HFDs) are related to metabolic syndrome, including abnormal blood lipid metabolism, elevated TG, elevated low-density lipoprotein, and hepatic lipid accumulation, leading to obesity, type 2 diabetes mellitus (T2DM), and cardiovascular disease (15, 16). Obesity is a continuously growing pandemic that can cause many diseases that affect quality of life (17). According to World Health Organization estimates, > 1.9 billion people aged ≥18 are overweight, and > 650 million of them are obese, which increases the risk of metabolic syndrome (18). Surgical intervention and the use of synthetic drugs for weight loss have limitations and side effects. For example, the anti-obesity drugs orlistat, topiramate, and sibutramine are available on the market, but they have severe side effects such as insomnia, anorexia, dry mouth, and gastrointestinal discomfort, which limit their use (19). Thus, it is key to identify new, effective, and easily tolerated anti-obesity agents from natural products (4, 2022).

NAFLD is a chronic metabolic stress-related liver injury closely related to genetics and IR; it is one of the clinical consequences of obesity (23). Serious consequences may lead to liver cirrhosis, hepatocellular carcinoma, coronary heart disease and diabetes, and steatosis increases inflammation (24). NAFLD is closely related to lifestyle such as HFD and diseases such as T2DM, dyslipidemia, adrenal hormone excess, obesity, and hypertension (25). The global incidence of NAFLD is increasing. Besides exercise and diet restrictions, there is currently a lack of effective treatment options (26, 27). G. cambogia had many activities related to metabolic syndrome because it was able to efficiently improve body fat mass, blood sugar level, body weight, TC, triglyceride level, glucose metabolism and display leptin-like activity (1, 28, 29), and its protective potential against NAFLD has been investigated (30). Despite these findings, to our knowledge the inhibitory effect of ethanol extract of G. cambogia in IR and NAFLD remain poorly understood. Therefore, this study explored the effect of GE, GED, and GEP on IR and hepatic lipid accumulation in HFD-induced obese mice.

Materials and methods

G. cambogia water extract

G. cambogia was purchased from Sri Lanka. The G. cambogia peel was subjected to extraction three times by placing it in boiling distilled water for 30 min each time. The solution was collected, concentrated, and lyophilized to obtain GE.

To obtain further extracts, GE and ethanol (1:3) were mixed together and then left to stand. Next, the supernatant was centrifuged. The resulting supernatant was designated the GED. After being left to stand, the precipitate was designated the GEP.

To assess the polysaccharides, total phenols, and flavonoids in GE, we used the phenol–sulfuric acid method, Folin–Ciocalteu method, and NaNO2-Al (NO3)3-NaOH colorimetric method, respectively. High-performance liquid chromatography (HPLC; Agilent 1200 HPLC, Jiangsu Jingmei Biotechnology Co., Ltd.) was used to assess the HCA levels in GE, GED, and GEP.

Animals and diets

Two animal experiments were conducted. In the first, HFD mice were treated with increasing concentrations of GE (0.2, 1, or 5 g/L GE) and compared to HFD-only control group. In the second experiment, HFD mice given GE, GED, or GEP were compared to HFD-only control group.

Male C57BL/6 mice aged 6–8 weeks were purchased from the Kunming Institute of Botany, Chinese Academy of Sciences and raised at Yunnan Agricultural University. During the entire experiment, the mice were kept in a specific-pathogen-free facility under a 12/12-h light/dark cycle and had free access to food and water (31). The mice were randomly divided into eight groups (six per group), and all were fed HFD (D12492, rodent diet with 60% kcal% fat, Research Diets, Inc.). The first four groups were as follows: HFD only, HFD+GE 0.2 g/L, HFD+GE 1 g/L, and HFD+GE 5 g /L. The second four groups were as follows: HFD only, HFD+GED 4 g/L, HFD+GEP 1 g/L, and HFD+GE 5 g/L. The corresponding extracts were added to the drinking water for 16 weeks. We recorded the weight and fasting blood glucose (FBG) of each mouse every week, as well as the changes in the amount of water and food intake of each mouse. After 16 weeks, the mice were sacrificed, and the liver, serum, inguinal fat, and epididymal fat were collected. All experimental procedures were performed in accordance with the guidelines specified by the Committee for Care and Use of Laboratory Animals of Yunnan Agricultural University and were approved by the Animal Experiments Ethics Committee of Yunnan Agricultural University (SCXY (Chuan) K2020-030) (32).

Testing tolerances for intraperitoneally injected glucose and insulin

In the 14th week, an intraperitoneal glucose tolerance test (IPGTT) was performed. The mice were fasted for 13 h, and the body weight of each mouse was measured and recorded. Each mouse was intraperitoneally injected with glucose solution (1 g/kg), and the blood glucose was measured at 0, 30, 60, 90, and 120 min with a blood glucose meter. The data were plotted, and the areas under the curve (AUCs) were calculated (33).

In the 15th week, an intraperitoneal insulin tolerance test (IPITT) was performed. The mice were not fasted, and the body weight of each mouse was measured and recorded. Insulin solution (0.75 U/kg) was injected into the intraperitoneal cavity of each mouse, and the blood glucose was measured at 0, 30, 60, 90, and 120 min using a blood glucose meter. The data were plotted, and the AUCs were calculated (34).

Cholesterol and aminotransferases

An Advia Chemistry XPT automatic biochemical analyzer (Siemens, Munich, Germany) at Kunming First People’s Hospital and Ganmei International Hospital was used to determine serum TC, high-density lipoprotein cholesterol (HDL-C), non-high-density lipoprotein cholesterol (non-HDL-C), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) levels.

Liver histological analysis

Mouse liver tissue was fixed with 4% paraformaldehyde, embedded in paraffin, subjected to hematoxylin and eosin (HE) staining, and fixed with neutral resin glue. The liver TG was measured using a TG kit (Nanjing Jiancheng Bioengineering Institute, Nanjing, China).

Statistical analysis

All data in this study are derived from three or more independent experiments. The data are expressed as the means ± standard deviation (SD). GraphPad Prism 9.0 software (GraphPad software, San Diego, CA, USA) was used to determine the statistical significance of differences between groups. The differences were assessed by one-way analysis of variance (ANOVA) and Tukey’s test. P < 0.05 was considered statistically significant.

Results

GE inhibits HFD-induced body weight gain and IR

C57BL/6 mice fed HFD were given 0, 0.2, 1, or 5 g /L G. cambogia extract (GE) in drinking water for 16 weeks. The mice fed HFD for 16 weeks gained body weight (Fig. 1A) and developed IR (Fig. 1E–H). GE effectively inhibited HFD-induced obesity, and the body weight decrease of the highest-dose group (5 g/L GE) compared to the HFD mice was significant from the 10th week (18.64% lower, P < 0.05; 24.67% reductions in the16th, P < 0.001, Fig. 1A). GE did not affect the food or water consumption of mice, indicating that the anti-obesity effect of GE was not caused by appetite suppression or a change in water consumption (Fig. 1C–D).

Fig 1
Fig. 1. GE reduces HFD-induced obesity and IR. (A) Weight changes of the four groups of HFD mice administered 0, 0.2, 1, or 5 g /L GE for 16 weeks. (B) Weekly fasting blood glucose. (C) Food intake. (D) Water intake. (E) IPGTT results. (F) IPITT results. (G) AUC values for IPGTT. (H) AUC values for IPITT. (I) Liver-to-body weight ratio. (J). Inguinal adipose tissue-to-body weight ratio. (K). Epididymal fat tissue-to-body weight ratio. Data are expressed as mean ± SD. One-way ANOVA and Tukey’s tests were used to analyze statistical differences. *P < 0.05, **P < 0.01, ***P < 0.001, ##P < 0.01. *HFD vs. HFD+5 g/L, #HFD vs. HFD+1 g/L.

Obesity is often accompanied by abnormal glucose metabolism. Therefore, we further assessed the effects of GE on glucose homeostasis by testing blood glucose levels using IPGTT and IPITT. The blood glucose levels of the HFD mice were elevated, while the highest-dose GE group exhibited significantly decreased FBG levels compared to the HFD mice (Fig. 1B). HFD mice exhibited a significant impairment in glucose tolerance and an increase in IR, as indicated by IPGTT and IPITT, respectively (Fig. 4E–H). GE increased glucose tolerance (a 37.97% reduction AUC in the 5 g/L GE) and decreased IR (a 31.78% reduction AUC in the 5 g/L GE) in HFD mice. GE significantly reduced the liver-to-body weight and inguinal fat-to-body weight ratios (18.64% lower) compared to those in the HFD group, and the high-dose GE group had a highly significant inhibitory effect on both (32.25% lower, P < 0.001; 51.46% lower, P < 0.01, respectively, Fig. 1I–J). However, GE had no significant effect on the epididymal fat-to-body weight ratio (Fig. 1K).

GE improves HFD-induced dyslipidemia and liver damage

Long-term HFD caused dyslipidemia in mice according to the blood test results. TC and non-HDL-C in the HFD group were abnormally increased by 23.41–39.51%, 57.33–68.40%, and GE significantly improved these factors (Fig. 2A and 2C). Long-term HFD also impaired liver function, which manifested as increased ALT, and 5g/L GE inhibited the HFD-induced increase in ALT (69.40% reduction, P < 0.01, Fig. 2D). However, GE did not significantly change the HDL-C or AST levels.

Fig 2
Fig. 2. GE reduces dyslipidemia. (A) TC. (B) HDL-C. (C) Non-HDL-C. (D) ALT. (E) AST. (F) ALT/AST. Data are expressed as mean ± SD. One-way ANOVA and Tukey’s tests were used to analyze statistical differences. *P < 0.05, **P < 0.01, ***P < 0.001.

Effect of GE on HFD-induced hepatic lipid accumulation

NAFLD is considered an important public health issue, but there is currently no effective treatment. To assess the effect of GE on HFD-induced hepatic lipid accumulation, mouse liver tissues were subjected to HE staining and TG assessment. GE inhibited the HFD-induced hepatic lipid accumulation (based on the images in Fig. 3A), and dose-dependently significantly inhibited the HFD-induced increase in TG (11.68–27.85%, Fig. 3B).

Fig 3
Fig. 3. GE improves HFD-induced hepatic lipid accumulation. (A) Liver morphology and HE staining in the four groups of HFD mice administered 0, 0.2, 1, or 5 g /L GE. (B) Hepatic TG level. Data are expressed as mean ± SD. One-way ANOVA and Tukey’s tests were used to analyze statistical differences. *P < 0.05, **P < 0.01, ***P < 0.001.

Fig 4
Fig. 4. GED and GEP alleviate HFD-induced obesity and IR. GED: 4 g/L, GEP: 1 g/L, GE: 5 g/L. (A) Weight change of the four groups of HFD mice over 16 weeks. (B) Weekly fasting blood glucose. (C) Food intake. (D) Water intake. (E) IPGTT results. (F) IPITT results. (G) AUC values for IPGTT. (H) AUC values for IPITT. (I) Liver-to-body weight ratio. (J) Inguinal adipose tissue-to-body weight ratio. (K) Epididymal fat tissue-to-body weight ratio. Data are expressed as mean ± SD. One-way ANOVA and Tukey’s tests were used to analyze statistical differences. *P < 0.05, **P < 0.01, ***P < 0.001, ##P < 0.01, +P < 0.05, ++P < 0.01. *HFD versus HFD+GE, #HFD versus HFD+GED, +HFD versus HFD+GEP.

GED and GEP inhibit HFD-induced obesity and IR

Next, we assessed the effects of the GED or precipitated (GEP) using ethanol. Long-term HFD led to obesity in the mice. Neither GED nor GEP reduced food or water consumption (Fig. 4C–D). The effect of GED regarding body weight reduction was highly significant (22.22–29.39% lower, P < 0.01) (Fig. 4A), but that of GEP was not significant. This may be due to the lower dose in the GEP group compared to the GED group (1 g/L vs. 4 g/L), and there was also a slightly lower HCA level in GEP compared to GED (19.79% vs. 22.81%).

To verify the effects of GED and GEP on IR, the mice were subjected to IPGTT and IPITT. The mice in the HFD group developed glucose intolerance and IR. GED and GEP alleviated both of these changes (Fig. 4E–H), and the AUCs for GED (but not GEP) were significantly lower compared to that for the HFD group (GTT and ITT were reduced by 28.49%, 32.07%, P < 0.05 and P < 0.01, respectively).

GED and GEP significantly inhibited the HFD-induced increase in liver weight by 29.43%, 29.34% (Fig. 4I, P < 0.01), and GED reduced inguinal fat (Fig. 4J). GED and GEP had no significant effect on FBG or epididymal fat in mice (Fig. 4B and K).

GED and GEP alleviate HFD-induced dyslipidemia and liver damage

The abnormal increases in dyslipidemia and liver damage markers in the HFD group (Fig. 5A–F) are predictors of atherosclerosis and coronary heart disease. TC, HDL-C, non-HCL-C, ALT, and ALT/AST levels were reduced by 25.61, 17.7737.7874.32, and 63.44%, respectively, in the mice fed HFD + GED than in the HFD mice (P < 0.05). However, GEP did not significantly inhibit HFD-induced increases in cholesterol (TC, HDL-C, and non-HCL-C) in mice; it only significantly inhibited ALT (52.43% reduction, P < 0.01) and ALT/AST (50.54% reduction, P < 0.01). This was potentially because the low dose of GEP compared to GED (1 g/L vs. 4 g/L) led to a lower HCA level, along with the slightly lower HCA level in GEP compared to GED (19.79% vs. 22.81%), so the effect was not as obvious.

Fig 5
Fig. 5. GED and GEP reduce dyslipidemia. GED: 4 g/L, GEP: 1 g/L, GE: 5 g/L. (A) TC. (B) HDL-C. (C) Non-HDL-C. (D) ALT. (E) AST. (F) ALT/AST. Data are expressed as mean ± SD. One-way ANOVA and Tukey’s tests were used to analyze statistical differences, *P < 0.05, **P < 0.01, ***P < 0.001.

GED and GEP inhibit HFD-induced hepatic lipid accumulation

HE staining and liver TG assessment showed that GED and GEP alleviated HFD-induced hepatic lipid accumulation (based on the images in Fig. 6A) and significantly inhibited the HFD-induced increase in TG (22.43% reductions in GED, P < 0.001; 9.41% reductions in GEP, P < 0.05, Fig. 6B). The effect of GED was more significant (P < 0.001 vs. P < 0.05), which might have been caused by the higher dose in the GED group compared to the GEP group (4 g/L vs. 1 g/L), and there was also a slightly higher HCA level in GED compared to GEP (22.81% vs. 19.79%).

Fig 6
Fig. 6. GED and GEP alleviate HFD-induced hepatic lipid accumulation. GED: 4 g/L, GEP: 1 g/L, GE: 5 g/L. (A) Liver morphology and HE staining in the four groups of HFD mice. (B) Liver TG level. Data are expressed as mean ± SD. One-way ANOVA and Tukey’s tests were used to analyze statistical differences. *P < 0.05, **P < 0.01, ***P < 0.001.

Levels of polysaccharides, total phenols, flavonoids, and HCA in G. cambogia extract

The contents of polysaccharides, polyphenols, and flavonoids in GE were measured to be 18.88–23.98%, 15.09–17.59%, and 18.39–18.89%, respectively, indicating that the level of polysaccharides in GE was slightly higher. To understand whether the effect of G. cambogia on weight, IR, and NAFLD is related to HCA, the HCA levels in GE, GED and GEP were measured by HPLC, and the results were 34.04–35.40%, 22.51–23.11%, and 19.29–20.29%. The level of HCA was the highest in GE, and the level of HCA in GED group was higher than that in GEP group.

Discussion

NAFLD is one of the most common chronic liver diseases. Thus, exploring how to treat NAFLD using dietary supplements is warranted. We found that (GE, GED, and GEP) alleviated HFD-induced IR and hepatic lipid accumulation in mice. Additionally, it inhibited the HFD-induced increased body weight and inguinal fat in mice, and it also affected the HFD-induced changes in blood test results in mice, especially the changed in TC and non-HDL-C. The HCA levels in GE, GED, and GEP were determined by HPLC, and we speculate that the effects of GE were caused by HCA.

Obesity is a global public health problem, and it is often associated with metabolic diseases such as IR, T2DM, NAFLD, coronary heart disease, and dyslipidemia (3537). Obesity, caused by fat accumulation, is a serious risk factor for so-called lifestyle-related diseases (38). G. cambogia/HCA products are widely marketed as dietary supplements, and G. cambogia has been shown to have positive effects regarding reducing body weight, lowering glucose levels, and inhibiting fat synthesis (3942). Most related studies have shown that G. cambogia/HCA can reduce weight gain (9, 4345) and visceral fat accumulation (46, 47). However, some studies have shown that HCA has no lasting beneficial effect on weight loss, which may be due to the short duration of the study (48). Our research showed that GE inhibited HFD-induced weight gain in mice, but it was not related to food intake (Figs. 1A, 1C, 4A, and 4C), which was different from previous studies showing that G. cambogia reduces body weight by suppressing appetite (7, 49). It also inhibited HFD-induced inguinal fat accumulation in mice, but it had no obvious effect on epididymal fat accumulation. Abnormal blood lipid metabolism plays a major role in the whole process of atherosclerosis occurrence, development, and deterioration, and is a major risk factor for coronary heart disease (41). We found that G. cambogia significantly reduced the HFD-induced mice serum increases in TC and non-HDL-C. In addition, we found that G. cambogia had a better effect on HFD-induced mice serum ALT than AST (Figs. 2D, 2E, 5D, and 5E), indicating that G. cambogia may inhibit HFD-induced hepatocyte membrane damage. These findings have potential significance for the treatment of metabolic disorders.

In addition to inducing weight loss, G. cambogia relieved IR and hepatic lipid accumulation. IR in metabolic diseases has received considerable attention in recent years (50). It is one of the key components of metabolic syndrome (51, 52). We found that G. cambogia alleviates HFD-induced glucose intolerance and IR (Figs. 1E–H and 4E–H), which is consistent with the results of previous studies (29, 53). It was recently reported that G. cambogia and HCA activated the antioxidant effect of Nrf2 antioxidant response element and improved NAFLD by regulating adipogenesis and apoptosis (54). Relatedly, using a mouse model of HFD-induced obesity, we found that G. cambogia dose-dependently improved hepatic lipid accumulation (Figs. 3 and 6), providing a new option for the treatment of NAFLD.

In conclusion, this study demonstrated that G. cambogia alleviates IR and hepatic lipid accumulation and has a weight-loss effect in HFD mice. These effects may be caused by the HCA in G. cambogia. Although we did not further assess whether HCA mediates the effects, based on our results, G. cambogia still has potential for the treatment of NAFLD.

Authors’ contributions

JS and CF received funding. CF conceived and designed the experiment. JD performed the experiments. JD, HZ, XD, XH, BD, WL, and YT contributed to the analysis and interpretation of the data. JD drafted the first edition of the manuscript, and CF edited the manuscript.

Ethics statement

The animal study was reviewed and approved by the Animal Experiment Ethics Review Committee of Yunnan Agricultural University.

Data availability statement

The original contributions presented in the study are included in the article; further inquiries can be directed to the corresponding author/s.

References

1. Fassina P, Scherer Adami F, Terezinha Zani V, Kasper Machado IC, Garavaglia J, Quevedo Grave MT, et al. The effect of Garcinia cambogia as co-adjuvant in the weight loss process. Nutr Hosp 2015; 32(6): 2400–8. doi: 10.3305/nh.2015.32.6.9587
2. Semwal RB, Semwal DK, Vermaak I, Viljoen A. A comprehensive scientific overview of Garcinia cambogia. Fitoterapia 2015;102:134–48. doi: 10.1016/j.fitote.2015.02.012
3. Han JH, Jang KW, Myung CS. Garcinia cambogia attenuates adipogenesis by affecting CEBPB and SQSTM1/p62-mediated selective autophagic degradation of KLF3 t-hrough RPS6KA1 and STAT3 suppression. Autophagy 2021; 8: 1–22. doi: 10.1080/15548627.2021.1936356
4. Attia RT, Abdel-Mottaleb Y, Abdallah DM, El-Abhar HS, El-Maraghy NN. Raspberry ketone and Garcinia cambogia rebalanced disrupted insulin resistance and leptin signaling in rats fed high fat fructose diet. Biomed Pharmacother 2019; 110: 500–9. doi: 10.1016/j.biopha.2018.11.079
5. Kim D, Kim J, Kim S, Yoon M, Um M, Kim D, et al. Arousal-inducing effect of Garcinia cambogia peel extract in pentobarbital-induced sleep test and eElectroencep-halographic analysis. Nutrients 2021; 13(8): 2845. doi: 10.3390/nu13082845
6. Lewis YS, Neelakantan S. (-)-Hydroxycitric acid – the principal acid in the fruits of Garcinia cambogia desr. Phytochemistry 1965; 4: 610–52. doi: 10.1016/s0031-9422(00)86224-x
7. Márquez F, Babio N, Bulló M, Salas-Salvadó J. Evaluation of the safety and efficacy of hydroxycitric acid or Garcinia cambogia extracts in humans. Crit Rev Food Sci Nutr 2012; 52(7): 585–94. doi: 10.1080/10408398.2010.500551
8. Chuah LO, Ho WY, Beh BK, Yeap SK. Updates on antiobesity effect of Garcinia origin (-)-HCA. Evid Based Complement Alternat Med 2013; 2013: 751658. doi: 10.1155/2013/751658
9. Kim KY, Lee HN, Kim YJ, Park T. Garcinia cambogia extract ameliorates visceral adiposity in C57BL/6J mice fed on a high-fat diet. Biosci Biotechnol Biochem 2008; 72(7): 1772–80. doi: 10.1271/bbb.80072
10. Loe YC, Bergeron N, Rodriguez N, Schwarz JM. Gas chromatography/mass spectrometry method to quantify blood hydroxycitrate concentration. Anal Biochem 2001; 292(1): 148–54. doi: 10.1006/abio.2001.5046
11. Li L, Zhang H, Yao Y, Yang Z, Ma H. (-)-Hydroxycitric acid suppresses lipid Droplet accumulation and accelerates energy metabolism via activation of the adiponectin-AMPK signaling pathway in broiler chickens. J Agric Food Chem 2019; 67(11): 3188–97. doi: 10.1021/acs.jafc.8b07287
12. Downs BW, Bagchi M, Subbaraju GV, Shara MA, Preuss HG, Bagchi D. Bioefficacy of a novel calcium-potassium salt of (-)-hydroxycitric acid. Mutat Res 2005; 579(1–2): 149–62. doi: 10.1016/j.mrfmmm.2005.02.021
13. 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(29): 4689–701. doi: 10.3748/wjg.v19.i29.4689
14. Vasques CA, Schneider R, Klein-Junior LC, Falavigna A, Piazza I, Rossetto S. Hypolipemic effect of Garcinia cambogia in obese women. Phytother Res 2014; 28(6): 887–91. doi: 10.1002/ptr.5076
15. Julibert A, Bibiloni MDM, Tur JA. Dietary fat intake and metabolic syndrome in adults: a systematic review. Nutr Metab Cardiovasc Dis 2019; 29(9): 887–905. doi: 10.1016/j.numecd.2019.05.055
16. Engin A. The definition and prevalence of obesity and metabolic syndrome. Adv Exp Med Biol 2017; 960: 1–17. doi: 10.1007/978-3-319-48382-5_1
17. Arrieta F, Pedro-Botet J. Recognizing obesity as a disease: a true challenge (Reconocer la obesidad como enfermedad: todo un reto). Rev Clin Esp (Barc) 2021; 221(9): 544–6. doi: 10.1016/j.rceng.2020.08.005
18. World Health Organization. Obesity and overweight. Key facts. 2021. [cited 2021 June 09]. Available from: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight
19. Derosa G, Cicero AF, Murdolo G, Piccinni MN, Fogari E, Bertone G, et al. Efficacy and safety comparative evaluation of orlistat and sibutramine treatment in hyperte-nsive obese patients. Diabetes Obes Metab 2005; 7(1): 47–55. doi: 10.1111/j.1463-1326.2004.00372.x
20. Yun JW. Possible anti-obesity therapeutics from nature – a review. Phytochemistry 2010; 71(14–15): 1625–41. doi: 10.1016/j.phytochem.2010.07.011
21. Feng Z, Chen J, Feng L, Chen C, Ye Y, Lin L. Polyisoprenylated benzophenone derivatives from Garcinia cambogia and their anti-inflammatory activities. Food Funct 2021; 12(14): 6432–41. doi: 10.1039/d1fo00972a
22. Vuppalanchi R, Bonkovsky HL, Ahmad J, Barnhart H, Durazo F, Fontana RJ, et al. Garcinia cambogia, either alone or in combination with green tea, causes moderate to severe liver injury. Clin Gastroenterol Hepatol 2022; 20(6): e1416–25. doi: 10.1016/j.cgh.2021.08.015
23. Fujii H, Kawada N, Japan Study Group of Nafld J-N. The role of insulin resistance and diabetes in nonalcoholic fatty liver disease. Int J Mol Sci 2020; 21(11): 3863. doi: 10.3390/ijms21113863
24. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease. J Hepatol 2016; 64: 1388–402. doi: 10.1016/j.jhep.2015.11.004
25. Nakajima T, Naito H. Mechanism analysis and prevention of pathogenesis of nona-lcoholic steatohepatitis. Nihon Eiseigaku Zasshi 2015; 70(3): 197–204. doi: 10.1265/jjh.70.197
26. Milic S, Mikolasevic I, Krznaric-Zrnic I, Stanic M, Poropat G, Stimac D, et al. Non-alcoholic steatohepatitis: emerging targeted therapies to optimize treatment options. Drug Des Devel Ther 2015; 9: 4835–45. doi: 10.2147/DDDT.S64877
27. Weiss J, Rau M, Geier A. Non-alcoholic fatty liver disease: epidemiology, clinical course, investigation, and treatment. Dtsch Arztebl Int 2014; 111(26): 447–52. doi: 10.3238/arztebl.2014.0447
28. Kuswandi A, Tarawaan VM, Goenawan H, Muchtaridi M, Lesmana R. Potential roles of Garcinia family as antimetabolic syndrome. J Adv Pharm Technol Res 2022; 13(1): 1–6. doi: 10.4103/japtr.japtr_218_21
29. Hayamizu K, Hirakawa H, Oikawa D, Nakanishi T, Takagi T, Tachibana T, et al. Effect of Garcinia cambogia extract on serum leptin and insulin in mice. Fitoterapia 2003; 74(3): 267–73. doi: 10.1016/s0367-326x(03)00036-4
30. Cha JY, Nepali S, Lee HY, Hwang SW, Choi SY, Yeon JM, et al. Chrysanthemum indicum L. ethanol extract reduces high-fat diet-induced obesity in mice. Exp Ther Med 2018; 15(6): 5070–6. doi: 10.3892/etm.2018.6042
31. Yang J, Ran Y, Yang Y, Song S, Wu Y, Qi Y, et al. 4-Hydroxyisoleucine alleviates macrophage-related chronic inflammation and metabolic syndrome in mice fed a high-fat diet. Front Pharmacol 2020; 11: 606514. doi: 10.3389/fphar.2020.606514
32. Fang C, Cai X, Hayashi S, Hao S, Sakiyama H, Wang X, et al. Caffeine-stimulated muscle IL-6 mediates alleviation of non-alcoholic fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864(3): 271–80. doi: 10.1016/j.bbalip.2018.12.003
33. Guan H, Wang Y, Li H, Zhu Q, Li X, Liang G, et al. 5-Bis-(2,6-difluoro-benzyliden-e) Cyclopentanone acts as a selective 11beta-hydroxysteroid dehydrogenase one inhibitor to treat diet-induced nonalcoholic fatty liver disease in mice. Front Pharmacol 2021; 12: 594437. doi: 10.3389/fphar.2021.594437
34. Ardaiz N, Gomar C, Vasquez M, Tenesaca S, Fernandez-Sendin M, Di Trani CA, et al. Insulin fused to apolipoprotein A-I reduces body weight and steatosis in DB/DB mice. Front Pharmacol 2020; 11: 591293. doi: 10.3389/fphar.2020.591293
35. Kyrou I, Randeva HS, Tsigos C, Kaltsas G, Weickert MO. Clinical problems caused by obesity. South Dartmouth, MA: MDText.com, Inc; 2000–2018.
36. Mir FA, Ullah E, Mall R, Iskandarani A, Samra TA, Cyprian F, et al. Dysregulated metabolic pathways in subjects with obesity and metabolic syndrome. Int J Mol Sci 2022; 23(17): 9821. doi: 10.3390/ijms23179821
37. Zaman CF, Sultana J, Dey P, Dutta J, Mustarin S, Tamanna N, et al. A Multidisciplinary approach and current perspective of nonalcoholic fatty liver disease: a systematic review. Cureus 2022; 14(9): e29657. doi: 10.7759/cureus.29657
38. Tchernof A, Després JP. Pathophysiology of human visceral obesity: an update. Physiol Rev 2013; 93(1): 359–404. doi: 10.1152/physrev.00033.2011
39. Sullivan AC, Triscari J, Hamilton JG, Miller ON. Effect of (-)-hydroxycitrate upon the accumulation of lipid in the rat. II. Appetite. Lipids 1974; 9(2): 129–34. doi: 10.1007/BF02532137
40. Sullivan AC, Triscari J, Hamilton JG, Miller ON, Wheatley VR. Effect of (-)-hydro-xycitrate upon the accumulation of lipid in the rat. I. Lipogenesis. Lipids 1974; 9(2): 121–8. doi: 10.1007/BF02532136
41. Sripradha R, Sridhar MG, Maithilikarpagaselvi N. Antihyperlipidemic and antioxidant activities of the ethanolic extract of Garcinia cambogia on high fat diet-fed rats. J Complement Integr Med 2016; 13(1): 9–16. doi: 10.1515/jcim-2015-0020
42. Han JH, Jang KW, Park MH, Myung CS. Garcinia cambogia suppresses adipogenes-is in 3T3–L1 cells by inhibiting p90RSK and Stat3 activation during mitotic clonal expansion. J Cell Physiol 2021; 236(3): 1822–39. doi: 10.1002/jcp.29964
43. Sripradha R, Magadi SG. Efficacy of Garcinia cambogia on body weight, inflammation and glucose tolerance in high fat fed male Wistar rRats. J Clin Diagn Res 2015; 9(2): BF01–4. doi: 10.7860/JCDR/2015/12045.5577
44. Maia-Landim A, Ramirez JM, Lancho C, Poblador MS, Lancho JL. Long-term effects of Garcinia cambogia/glucomannan on weight loss in people with obesity, PLI-N4, FTO and Trp64Arg polymorphisms. BMC Complement Altern Med 2018; 18(1): 26. doi: 10.1186/s12906-018-2099-7
45. Bilal T, Gursel FE, Ates A, Altiner A. Effect of Garcinia cambogia extract on body weight gain, feed intake and feed conversion ratio, and serum non-esterified fatty acids and C-reactive protein levels in rats fed with atherogenic diet. Iran J Vet Res 2012; 13(4): 330–3. doi: 10.22099/IJVR.2012.614
46. Clouatre DL, Preuss HG. Hydroxycitric acid does not promote inflammation or liver toxicity. World J Gastroenterol 2013; 19(44): 8160–2. doi: 10.3748/wjg.v19.i44.8160
47. Hayamizu K, Ishii Y, Kaneko I, Shen M, Okuhara Y, Shigematsu N, et al. Effects of garcinia cambogia (Hydroxycitric Acid) on visceral fat accumulation: a double-blind, randomized, placebo-controlled trial. Curr Ther Res Clin Exp 2003; 64(8): 551–67. doi: 10.1016/j.curtheres.2003.08.006
48. Onakpoya I, Hung SK, Perry R, Wider Barbara, Ernst E. The use of Garcinia extract (hydroxycitric acid) as a weight loss supplement: a systematic review and meta-analysis of randomised clinical trials. J Obes 2011; 2011: 509038. doi: 10.1155/2011/509038
49. Han N, Li L, Peng M, Ma H. (-)-Hydroxycitric acid nourishes protein synthesis via altering metabolic directions of amino acids in male rats. Phytother Res 2016; 30(8): 1316–29. doi: 10.1002/ptr.5630
50. Hill MA, Yang Y, Zhang L, Sun Z, Jia G, Parrish AR, et al. Insulin resistance, cardiovascular stiffening and cardiovascular disease. Metabolism 2021; 119: 154766. doi: 10.1016/j.metabol.2021.154766
51. Zagayko AL, Shkapo AI, Fylymonenko VP, Briukhanova TO. The impact of hydroxycitric acid on the lipid metabolism profile under experimental insulin resistance syndrome of Syrian hamsters. Ukr Biochem J 2016; 88(3): 78–82. doi: 10.15407/ubj88.03.078
52. Gluvic Z, Zaric B, Resanovic I, Obradovic M, Mitrovic A, Radak D, et al. Link between metabolic syndrome and insulin resistance. Curr Vasc Pharmacol 2017; 15(1): 30–9. doi: 10.2174/1570161114666161007164510
53. Amin KA, Kamel HH, Abd Eltawab MA. The relation of high fat diet, metabolic di-sturbances and brain oxidative dysfunction: modulation by hydroxy citric acid. Lipids Health Dis 2011; 10: 74. doi: 10.1186/1476-511X-10-74
54. Han JH, Park MH, Myung CS. Garcinia cambogia ameliorates non-alcoholic fatty liver disease by inhibiting oxidative stress-mediated steatosis and apoptosis through NRF2-ARE activation. Antioxidants (Basel) 2021; 10(8): 1226. doi: 10.3390/antiox10081226