Journal List > Nutr Res Pract > v.15(3) > 1147158

Kim, Kim, and Cha: The protective effects of steamed ginger on adipogenesis in 3T3-L1 cells and adiposity in diet-induced obese mice

Abstract

BACKGROUND/OBJECTIVES

The steamed ginger has been shown to have antioxidative effects and a protective effect against obesity. In the present study, we investigated the effects of ethanolic extract of steamed ginger (SGE) on adipogenesis in 3T3-L1 preadipocytes and diet-induced obesity (DIO) mouse model.

MATERIALS/METHODS

The protective effects of SGE on adipogenesis were examined in 3T3-L1 adipocytes by measuring lipid accumulations and genes involved in adipogenesis. Male C57BL/6J mice were fed a normal diet (ND, 10% fat w/w), a high-fat diet (HFD, 60% fat w/w), and HFD supplemented with either 40 mg/kg or 80 mg/kg of SGE for 12 weeks. Serum chemistry was measured, and the expression of genes involved in lipid metabolism was determined in the adipose tissue. Histological analysis and micro-computed tomography were performed to identify lipid accumulations in epididymal fat pads.

RESULTS

In 3T3-L1 cells, SGE significantly decreased lipid accumulation, with concomitant decreases in the expression of adipogenesis-related genes. SGE significantly attenuated the increase in body, liver, and epididymal adipose tissue weights by HFD. Serum total cholesterol and triglyceride levels were significantly lower in SGE fed groups compared to HFD. In adipose tissue, SGE significantly decreased adipocyte size than that of HFD and altered adipogenesis-related genes.

CONCLUSIONS

In conclusion, steamed ginger exerted anti-obesity effects by regulating genes involved in adipogenesis and lipogenesis in 3T3-L1 cell and epididymal adipose tissue of DIO mice.

INTRODUCTION

Obesity is defined as the abnormal or excessive accumulation of fat in the body from the World Health Organization (WHO). It is a significant risk factor for metabolic diseases such as cardiovascular disease, nonalcoholic fatty liver disease, hypertension, type 2 diabetes, and a specific type of cancer [1]. Adipose tissue, a critical organ for energy homeostasis, expands to store excess energy in obesity [23]. Adipocytes, the primary cell type in adipose tissue, play a crucial role in adipose tissue enlargement [4]. Adipose tissue enlarges by increases of preexisting mature adipocytes in size (hypertrophy) and/or of numbers of new adipocytes (hyperplasia) [56]. Hypertrophy appears in the progression of obesity to meet the need for additional fat storage and then trigger hyperplasia in adipose tissue [25]. The enlargement of adipocytes arises from increased lipogenesis to promote lipid storage [7]. Adipogenesis, a process of preadipocytes differentiate into mature adipocytes, plays a crucial role in obesity. The process is tightly regulated by a network of transcription factors that activate a large number of adipogenesis genes [89]. The key players in the transcriptional cascade of adipocyte differentiation are peroxisome proliferator-activated receptor γ (PPARγ) and members of CCAAT/enhancer-binding protein (C/EBP) family [810]. PPARγ and C/EBP are the master regulators of adipogenesis and induce the genes involved in lipogenesis, lipolysis, and insulin sensitivity, such as glucose transporter 4 (GLUT4), adipocyte protein 2 (aP2), lipoprotein lipase (LPL), etc. [49]. Therefore, the inhibition of adipogenesis and triglyceride (TG) accumulation in the adipocytes is one of the winning strategies for the prevention of obesity [11].
Herbal medicines are claimed to effectively control obesity and its associated metabolic diseases [1213]. Ginger (Zingiber officinale) is one of the most commonly used spices and traditional medicine to treat arthritis, sprains, muscular pains, sore throats, nausea, and stroke in Asian countries [1415]. Ginger contains several bioactive phenolic compounds, including non-volatile and pungent compounds such as gingerol, shogaol, paradol, and zingerone [16]. The potential effects and its mechanisms actions of ginger and its active component, i.e., gingerol, shogaol, zingerone, etc., on the prevention of obesity have been reported in cell and animal studies [1415].
The composition and its functional properties of herbs or vegetables can differ from cultivation, storage, and processing methods. Ginger is heated or steamed when used as a spice or traditional medicine. Several studies reported that the heating or steaming process of herbs or root vegetables changes the chemical profiles [1718], and these changes can affect or improve their bioactivities [19202122]. Steamed ginger exerted different chemical profiles with enhanced anticancer effect [23] and adrenaline secretion [24]. In the previous study, the ethanolic extract of steamed ginger (SGE) showed antioxidative activity and lowered serum lipids in obesity induced by a high-fat diet (HFD) in C57BL/6J mice [25]. However, little is known about the protective effects and its underlying mechanisms of steamed ginger on adipogenesis in adipocytes and diet-induced obese mice. In the present study, we investigated the anti-obesity effect and its underlying mechanisms of the steamed ginger on 3T3-L1 cells and C57BL/6J mice fed a HFD.

MATERIALS AND METHODS

Ginger extract preparation

The standardized SGE was prepared from the Healthy Local Food Branding Agency (Wanju, Korea) as previously described. The 6-shogaol served as a marker and active compound for the standardization, and the contents in the SGE was analyzed using HPLC at a detection wavelength of 230 nm [2526].

Cell culture and viability assay

The 3T3-L1 cells murine preadipocytes (ATCC, Manassas, VA, USA) were cultured in DMEM (Lonza, Walkersville, MD, USA) supplemented with 10% fetal bovine serum (Gibco, Grand Island, NY, USA), 100 U/mL of penicillin, 100 μg/mL of streptomycin (Hyclone, Logan, UT, USA) in a 37°C humidified cell culture incubator with 5% CO2. The cytotoxicity of SGE was assessed with the cell viability of 3T3-L1 preadipocyte cells. The cells were seeded at a final concentration of 1 × 104 and treated with different concentrations of SGE for 24, 48, and 72 h. After incubation, the MTT [3(4,5-dimetyl2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide] EZ-Cytox, Daeil Lab Service, Seoul, Korea] and incubated for 2 h and read on the absorbance at 450 nm using a microplate reader (MRX II, Dynex Technologies, Chantilly, VA, USA).

Cell Differentiation and SGE treatment

Two days after post-confluency, the preadipocytes were differentiated in DMEM supplemented with 10% FBS and differentiation cocktail containing 0.5 mM IBMX, 1 μM DEXA, and 10 μg/mL insulin (Sigma-Aldrich Co., St. Louis, MO, USA). After differentiation, the cells were maintained in 10% FBS and 10 μg/mL of insulin in DMEM for another 6 days. 90% of the cells had transformed into mature adipocytes, with the formation of intracellular lipid droplets. SGE was treated every other day during the differentiation period of 8 days.

Oil-red O staining and glycerol-3-phosphate dehydrogenase (GPDH) assay

The cells were stained with Oil-Red O solution (Sigma-Aldrich Co.) to determine oil droplets in adipocytes as previously described [27]. Briefly, the differentiated cells were fixed with 10% formalin and then stained with filtered Oil-Red O solution for 30 min. The cellular TG contents were measured by the cell lysate (5% Triton X-100) at 580 nm. The GPDH activity was determined using GPDH assay kits from Takara (Takara Bio Inc., Otsu, Japan).

Animal care and diet

Eight-week-old male C57BL/6J mice (Charles River Laboratories, Tokyo, Japan) were randomly divided into four groups (n = 10 per group, total n = 40): a normal diet (ND; 10% fat by wt, Research Diets, InC., NJ, USA), a HFD (60% fat by wt, Research Diets, Inc.), and HFD supplemented with 40 mg/kg or 80 mg/kg of SGE by wt (SGD4 or SGD8, respectively). All animal procedures were approved by the Institutional Animal Care and Use Committee at the Chonbuk National University (CBU 2016). The mice were housed in a polycarbonate cage under 12 h light/dark cycles. The composition of the experimental diets fed to the mice for 12 weeks are shown in Table 1. Body weight and food consumption were recorded weekly. After 12 weeks, mice were fasted for 12 h and sacrificed. The serum was collected by centrifugation of blood at 1,000 × g for 15 min at 4°C. The epididymal adipose fat pads were snap-frozen in liquid nitrogen or fixed in 10% formalin. The serum and tissue samples were stored at −80°C until use.
Table 1

Composition of experimental diets (unit: g)

nrp-15-279-i001
Component (100 g−1) ND HFD
Casein 18.96 25.84
Cystine 0.28 0.39
Corn starch 29.86 -
Maltodextrin 3.32 16.15
Sucrose 33.17 8.89
Cellulose 4.74 4.46
Soybean oil 2.37 3.23
Lard 1.90 31.66
Mineral 0.95 1.29
Dicalcium phosphate 1.23 1.68
Calcium carbonate 0.52 0.71
Potassium citrate 1.56 2.13
Vitamin mix 0.95 1.29
Choline bitartrate 0.19 0.26
Kcal g−1 3.85 5.24
ND: 10% of energy as fat (D12450B), HFD: 60% of energy as fat (D12492).
ND, normal diet; HFD, high-fat diet.

Serum chemistry and liver lipid measurements

Total cholesterol (TC) and TG concentrations in serum were determined by a commercial assay kit (Asan Pharmaceutical Co., Seoul, Korea). Serum alanine transaminase (ALT) and aspartate transferase (AST) activity were measured using a chemistry analyzer (Fuji Dri-Chem 3500, Fuji Photo Film Co., Tokyo, Japan). Serum glucose levels and insulin were measured by glucometer (Accu-check, Roche, Mannheim, Germany) and insulin ELISA kit (Alpeo, Sale, MA, USA), respectively. Leptin and adiponectin concentrations in serum were analyzed by assay kits (R&D Systems, Minneapolis, MN, USA), followed by the manufacturer's protocols. Hepatic TC and TG were measured enzymatically measured from the lipids extracted by Folch's methods, as we previously described [27].

Quantification of abdominal fat using micro-computed tomography (micro-CT)

The abdominal fat was imaged using high-resolution in vivo micro-CT (Skyscan, Konitch, Belgium) as previously described [2527]. The abdominal micro-CT scan images of anesthetized mice were captured at the level of L1-L5 intervertebral disk and analyzed using CT analyzer Version 1.16.4.1, Skyscan software (Skyscan).

Histology of epididymal adipose tissue

The formalin-fixed epididymal adipose tissue samples were embedded in paraffin. The paraffin-embedded tissues were cut at a thickness of 5 sµm and stained with hematoxylin-eosin. The tissue images were viewed using an Axiophot Zeiss Z1 microscope (Carl Zeiss, Gottingen, Germany) and analyzed with Analysis SIS3.2 software (Soft-Imaging System).

Gene expression analysis by quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA was extracted using TRIzol reagent (Life Technologies, Inc., Carlsbad, CA, USA), and qRT-PCR analysis for gene expression was conducted as previously described using the SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA, USA) and 7500 real-time PCR system (Applied Biosystems) [2829]. The primer sequences for qRT-PCR used in the present study are listed in Table 2. The glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used as an internal control.
Table 2

Quantitative real-time polymerase chain reaction primer

nrp-15-279-i002
Gene Primers Sequence (5′-3′)
PPARγ Forward GTGCCAGTTTCGATCCGTAGA
Reverse GGCCAGCA TCGTGTAGATGA
C/EBPα Forward GTGTGCACGTCTATGCTAAACCA
Reverse GCCGTTAGTGAAG AGTCTCAGTTT
aP2 Forward CTCGAAGGTTTACAAAATGTGTGA
Reverse AAACTCTTGTGGAAGTCACGCCTTT
GLUT4 Forward ACCGAGACAGGCCTCAGTGTG
Reverse GAATCGGCCACCGGTAAAGAG
FAS Forward AGGGGTCGACCTGGTCCTCA
Reverse GCCATGCCCAGAGGGTGGTT
ACC Forward CCAACATGAGGACTATAACTTCCT
Reverse TACATACGTGCCGTCAGGCTTCAC
ApN Forward GCACTGGCAAGTTCTACTGCAA
Reverse GTAGGTGAAGAGAACGGCCTTGT
ATGL Forward AACACCAGCATCCAGTTCAA
Reverse GGTTCA GTAGGCCATTCCTC
HSL Forward ACCGAGACAGGCCTCAGTGTG
Reverse GAATCGGCCACCGGTAAAGAG
SREBP1c Forward ACCGAGACAGGCCTCAGTGTG
Reverse GAATCGGCCACCGGTAAAGAG
PPARγ, peroxisome proliferator activated receptor gamma; C/EBPα, CCAAT/enhancer binding protein alpha; aP2, adipocyte protein 2; GLUT4, glucose transporter 4; FAS, fatty acid synthase; ACC, acetyl CoA carboxylase; ApN, adiponectin; ATGL, adipose triglyceride lipase; HSL, hormone sensitive lipase; SREBP1c, sterol regulatory element-binding protein1c.

Statistical analysis

One-way analysis of variance (ANOVA) and Newman-Keuls post hoc analysis was performed to detect the significance of mean differences between groups using GraphPad Prism 7 (GraphPad Software, Inc., La Jolla, CA, USA). The α-level of P < 0.05 was considered statistically significant, and all data are expressed as means ± SEM.

RESULTS

Inhibitory effect of SGE on lipid accumulation in 3T3-L1 adipocytes

The safety of SGE was considered by measuring cytotoxicity levels before the differentiation of preadipocytes. The cell viability of 3T3-L1 cells determined the concentrations of SGE. The cells were incubated with increasing concentrations (25 μg/mL) of SGE for 24, 48, and 72 h. There was no significant reduction in the cell viability by SGE at the range under 10 μg/mL (Fig. 1A). Therefore, the following experiments were done with a concentration of fewer than 10 μg/mL of SGE on 3T3-L1 cells. The increased lipid accumulation in adipocytes is a critical feature in adipogenesis. The contents of TG were measured to investigate whether SGE can attenuate lipid accumulation induced during 3T3-L1 differentiations. SGE significantly decreased the TG contents in the cells compared to control (Fig. 1B). Furthermore, SGE reduced the lipid droplet size and suppressed oil droplet accumulation on differentiated cells (Fig. 1C). The activity of GPDH, a representative marker for adipocyte differentiation, was significantly decreased in SGE treated cells (Fig. 1D).
Fig. 1

Effects of SGE on (A) cell viability (B) Triglyceride contents (C) Oil-Red O staining and (D) GPDH activity in 3T3-L1 cells. Cells were treated with different concentrations of SGE. Quantitative real-time-polymerase chain reaction analysis was conducted to measure mRNA abundance. Data are expressed as relative expression to GAPDH control. Bars with different letters are significantly different (P < 0.05). Values are means ± SEM; n = 6.

SGE, ethanolic extract of steamed ginger; GPDH, glycerol-3-phosphate dehydrogenase; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.
nrp-15-279-g001

Effect of SGE on the gene expression in 3T3-L1 adipocytes

To investigate the underlying mechanisms of SGE on 3T3-L1 adipocyte differentiation, we measured the genes involved in adipogenesis and lipogenesis. The expression of PPARγ, the master transcription factor for adipocyte differentiation, was significantly and dose-dependently decreased by SGE. There was a marked reduction of C/EBPα, a well-known regulator for adipogenesis, in SGE treated 3T3-L1 cells. During adipocyte differentiation, the expressions of aP2, GLUT4, fatty acids synthase (FAS), acetyl-CoA carboxylase (ACC), adiponectin (ApN) are regulated by the actions of PPARγ and C/EBPα. The mRNA abundance of aP2, GLUT4, FAS, ACC, and ApN was significantly decreased by SGE treatment. We also measured genes for lipolysis in SGE treated mature adipocytes (Fig. 2).
Fig. 2

Effects of SGE during differentiation of 3T3-L1 cells. Cells were treated with 1 or 5 μg/mL of SGE during differentiation. Quantitative real-time-polymerase chain reaction analysis was conducted to measure mRNA abundance. Data are expressed as relative expression to GAPDH control. Bars with different letters are significantly different (P < 0.05). Values are means ± SEM; n = 6.

SGE, ethanolic extract of steamed ginger; PPARγ, peroxisome proliferator-activated receptor gamma; C/EBPα, CCAAT/enhancer binding protein alpha; aP2, adipocyte protein 2; GLUT4, glucose transporter 4; FAS, fatty acid synthase; ACC, acetyl CoA carboxylase; ApN, adiponectin; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.
nrp-15-279-g002

Effect of SGE on body and tissue weights and serum chemistry in HFD-induced obese mice

The inhibitory effect of SGE on adipocyte differentiation in 3T3-L1 preadipocytes suggested the anti-obesity effect of SGE. We next examined the effects of SGE on DIO mice. The HFD significantly increased body, epididymal adipose fat, and mesenteric adipose fat weight compared to ND, whereas SGE supplementation significantly attenuated the increases. The increased liver weight and the elevation of hepatic TC and TG concentrations were observed in HFD-fed groups. However, SGE supplementation (40 mg/kg or 80 mg/kg) significantly attenuated the weight gain and the hepatic TC and TG concentrations compared to those of HFD (Table 3).
Table 3

Body weight, adipose weight, liver weight and lipid contents of C57BL/6J mice fed high-fat diet supplemented with SGE for 12 weeks

nrp-15-279-i003
Group Final body weight (g) Epididymal adipose weight (g) Mesenteric adipose weight (g) Liver weight (g) Liver TC (mg/g) Liver TG (mg/g)
ND 24.7 ± 0.3c 0.36 ± 0.03c 0.10 ± 0.01c 1.00 ± 0.04c 4.2 ± 0.3b 9.6 ± 1.5b
HFD 39.7 ± 0.5a 2.80 ± 0.08a 0.61 ± 0.04a 1.24 ± 0.06a 9.5 ± 1.3a 14.8 ± 1.0a
SGD4 37.1 ± 0.7b 2.54 ± 0.06b 0.49 ± 0.05b 1.15 ± 0.04b 6.8 ± 0.5b 10.6 ± 1.5b
SGD8 36.0 ± 0.9b 2.33 ± 0.12b 0.41 ± 0.05b 1.10 ± 0.06b 6.3 ± 2.6b 10.2 ± 0.4b
Data represent mean ± SEM (n = 10). Values with different letter in a column are significantly different (P < 0.05).
SGE, ethanolic extract of steamed ginger; TC, total cholesterol; TG, triglyceride; ND, normal diet; HFD, high-fat diet; SGD4, HFD with 40 mg/kg of SGE; SGD8, HFD with 80 mg/kg of SGE.
Serum TC and TG levels were significantly elevated in HFD compared to ND, whereas SGE significantly and dose-dependently attenuated the increases. Elevated serum glucose and insulin levels in the HFD groups, compared with the ND, were significantly (P < 0.05) decreased in SGE supplemented groups. Furthermore, the increased levels of serum adiponectin and leptin were markedly attenuated in SGD4 and SGD8 groups compared to those of HFD. Moreover, ALT and AST were significantly decreased in the serum of SGE supplemented DIO mice (Table 4).
Table 4

Serum chemistry of C57BL/6J mice fed high-fat diet supplemented with SGE for 12 week

nrp-15-279-i004
Parameters ND HFD SGD4 SGD8
TC (mg/dL) 109.5 ± 3.2d 161.3 ± 3.6a 143.3 ± 1.8b 131.1 ± 6.3c
TG (mg/dL) 40.7 ± 0.9c 78.6 ± 2.1a 65.4 ± 1.7b 63.9 ± 2.6b
Glucose (mg/dL) 65.4 ± 3.3c 102.1 ± 4.2a 97.1 ± 1.0ab 92.3 ± 1.4b
Insulin (ng/mL) 66.3 ± 2.9c 108.3 ± 5.9a 91.1 ± 3.5b 79.2 ± 5.6bc
Adiponectin (μg/mL) 8.1 ± 0.2c 15.7 ± 0.4a 13.1 ± 0.3b 12.8 ± 0.5b
Leptin (ng/mL) 40.7 ± 0.9c 78.6 ± 2.1a 65.4 ± 1.7b 63.9 ± 2.6b
ALT (units/L) 6.1 ± 0.1c 10.1 ± 0.6a 7.6 ± 0.3b 7.6 ± 0.2b
AST (units/L) 27.7 ± 0.8b 35.7 ± 2.5a 28.8 ± 1.5b 28.9 ± 1.2b
Data represent mean ± SEM (n = 10). Values with different letter in a raw are significantly different (P < 0.05).
SGE, ethanolic extract of steamed ginger; ND, normal diet; HFD, high-fat diet; SGD4, HFD with 40 mg/kg of SGE; SGD8, HFD with 80 mg/kg of SGE; TC, total cholesterol; TG, triglyceride; ALT, alanine transaminase; AST, aspartate transferase.

Effect of SGE on the epididymal adipose tissue of DIO mice

The effects of SGE on adipogenesis in the epididymal adipose tissue of DIO mice were examined by histology and micro-CT images. SGE supplementation attenuated the size and length of the adipocyte in epididymal adipose tissue compared to those of HFD (Fig. 3A and 3B). The micro-CT also showed that SGE significantly decreased the fat mass in HFD induced obesity (Fig. 3C and D).
Fig. 3

Epididymal adipose tissue in SGE supplemented diet-induced obesity mice for 12 weeks. (A) Hematoxylin and eosin stained adipose tissue (B) Adipocyte number (C) The representative images of micro-CT (D) The fat volume of mice using in vivo micro-CT image analysis. Bars with different letters are significantly different (P < 0.05). Values are means ± SEM; n = 10.

SGE, ethanolic extract of steamed ginger; ND, normal diet; HFD, high-fat diet; SGD4, HFD with 40 mg/kg of SGE; SGD8, HFD with 80 mg/kg of SGE; CT, computed tomography.
nrp-15-279-g003

Effect of SGE on the mRNA expression in epididymal adipose tissue of DIO mice

We investigated whether the alterations in the expressions of genes involved in adipogenesis and TG synthesis are responsible for decreasing weight and adipocyte size in epididymal adipose tissue of DIO mice by SGE supplementation. Interestingly, the mRNA abundance of transcription factors for adipogenesis and lipogenesis, i.e., PPAR, C/EBPα and sterol regulatory eleme0nt-binding protein1c (SREBP1c), was significantly reduced in epididymal adipose tissue of SGE groups compared to HFD. HFD exerted the decreased expression of GLUT4 and ApN in the epididymal adipose tissue, whereas the expressions were increased in both SGD4 and SGD8 groups. Furthermore, the increased expression of aP2, FAS, ACC in the adipose tissue of HFD were significantly decreased by SGE supplementation. In contrast, the expression of genes for lipolysis, ATGL and HSL, were significantly induced in the epididymal fat by SGE supplementation (Fig. 4A and B).
Fig. 4

Effect of SGE in the epididymal adipose tissue of mice. mRNA expression of gene involved in (A) adipogenesis and (B) lipid metabolism. Bars with different letters are significantly different (P < 0.05). Values are means ± SEM; n = 10.

SGE, ethanolic extract of steamed ginger; ND, normal diet; HFD, high-fat diet, SGD4, HFD with 40 mg/kg of SGE; SGD8, HFD with 80 mg/kg of SGE; PPARγ, peroxisome proliferator-activated receptorγ; C/EBPα, CCAAT/enhancer-binding proteinα; aP2, adipocyte protein 2; GLUT4, glucose transporter type 4; ApN, adiponectin; SREBP1c, sterol regulatory element-binding protein1c; FAS, fatty acid synthase; ACC, acetyl-coA carboxylase; ATGL, adipose triglyceride lipase; HSL, hormone-sensitive lipase.
nrp-15-279-g004

DISCUSSION

Obesity and its associated metabolic diseases are the major health problem worldwide, and herbal medicines are claimed to control these diseases effectively. Ginger, one of the widely used spices and traditional medicines, is on the United States Food and Drug Administration (FDA)'s Generally Recognized as Safe (GRAS) list. The beneficial effects and mechanisms of ginger and/or its bioactive phenolic components on obesity are well-known. However, most of the studies focused on the effects of raw, dried, or frozen ginger. Ginger may undergo a heating or steaming process when used as a spice or traditional medicine. The steaming process changed the chemical profiles of root vegetables and herbs, i.e., Panax notoginseng, Codonopsis lanceolate and garlic, etc., [1718] and affected their bioactivities [19202122]. Furthermore, Cheng et al. [23] reported that the steaming process affected the chemical profile of ginger, and the steamed ginger exerted a higher antiproliferative effect than that of dried and fresh ginger in Hela cells. The effects of ginger on obesity are well-known. However, to our knowledge, the present study is the first study investigating whether steamed ginger affects adipogenesis and adiposity in both cell and epididymal adipose tissue of the DIO mouse model.
In the present study, we investigated the effect of steamed ginger on adipogenesis in adipocytes and the DIO mice model. Adipogenesis and TG accumulations in adipocytes are associated with obesity and are essential to avoid ectopic fat deposition in non-adipose tissue [30]. In obesity, adipose tissue grows by hyperplasia (cell number increase) and/or hypertrophy (cell size increase). One winning strategy for the prevention of obesity is targeting adipogenesis [31].
Consistent with our study, the bioactive component of gingers showed an inhibitory effect on adipogenesis and lipid accumulation in 3T3-L1 cells [32333435363738]. The GPDH plays a significant role in TG accumulation in adipocytes by providing the glycerol for the TG synthesis. The phytochemicals of ginger, i. e., 6-shogaol, 6-gingerol, galanolactone, and gingerenone A, exerted inhibitory effects on intracellular lipid contents and GPDH activity in 3T3-L1 cells. In the present study, SGE significantly suppressed TG content and GPDH activity in 3T3-L1 cells. Adipogenesis is strictly regulated by the master switch of crucial transcription factors that activate adipogenesis genes [3940]. PPARγ and C/EBPα, the master regulators among the cascade of adipogenic transcription factors, are involved in the final stage of adipogenesis. These transcription factors induce their downstream genes that promote preadipocyte differentiation into mature adipocytes [394041]. Activation of PPARγ by its ligands also leads to adipocyte differentiation [42]. PPARγ alone or with C/EBPα induces genes involved in lipid storage, lipogenesis, and insulin sensitivity, e.g., aP2, cluster of differentiation 36 (CD36), LPL, GLUT4, and ApN [39404142]. SREBP-1c is the key transcription factor for lipogenesis and induces enzymes involved in lipogenesis such as ACC, FAS, and stearoyl-CoA desaturase-1 (SCD-1) [43]. The transcription factor also stimulates C/EBPα and PPARγ and plays an important role in adipogenesis [41]. SGE significantly inhibited adipocyte differentiation and lipid accumulation in 3T3-L1 cells by altering adipogenesis-related genes. The expressions of PPARγ and C/EBPα significantly, and dose-dependently decreased in SGE treated 3T3-L1 cells. Several studies reported that 6-gingerol, the primary active component of ginger, significantly reduced both mRNA and protein levels of PPARγ and C/EBPα with concomitant decreased adipogenesis-related genes during differentiation. [3334]. 6-gingerol inhibited adipocyte differentiation and cytoplasmic lipid accumulation by regulating the expression of adipogenesis-related genes via activating Wnt/b-catenin signaling pathway [35], or by inhibiting Akt/GSK3β-mediated signaling that suppresses the signaling cascades of PPARγ and C/EBPs during differentiation of 3T3-L1 cells [33]. Compared to 6-gingerol, 6-shogaol, another major active component of ginger, exerted higher inhibitory effects on the adipogenic process of 3T3-L1 preadipocytes by suppressing adipogenic/lipogenic marker proteins [36]. 6-shogaol and its major metabolite, 6-paradol, showed potent activity in stimulating glucose utilization by 3T3-L1 cells by increasing 5′ adenosine monophosphate-activated protein kinase (AMPK) phosphorylation [37]. Galanolactone isolated from ginger exerted a significant reduction in fat accumulation in adipocytes with downregulation of PPARγ and C/EBPα and aP2 mRNA expression [38]. The major bioactive components of ginger are 6-gingerol and 6-shogaol [44]. SGE contained the highest contents of 6-shogaol, among other extracts (data not shown). Therefore, these active compounds present in SGE may have potent inhibitory effects on adipocyte differentiation by altering genes involved in adipogenesis and lipogenesis.
In 3T3-L1 cells, SGE exerted strong inhibitory effects on differentiation by regulating adipogenesis related gene. Therefore, we next investigated whether SGE can have protective effects on DIO mice. In the present study, HFD significantly increased body weight gain, weights of epididymal adipose tissue, mesenteric adipose tissue, and liver compared to those of ND in C57BL/6J mice. Furthermore, HFD induced the elevation of serum TC and TG levels compared to those of ND. However, SGE supplementation (40 mg/kg or 80 mg/kg) significantly attenuated the weight gains and the increased serum TC and TG levels compared to those of HFD. Consistent with our results, studies reported the hypolipidemic and hypocholesterolemic effects of ethanolic extracts of ginger by improving TC, LDL-C, and TG levels in high-fat DIO C57BL/6J mice [45], apolipoprotein E-deficient mice [46], and goldthioglucose-induced obese mice [47]. Furthermore, ethanolic extract of ginger exerted hypolipidemic and hypocholesterolemic effects in several animal models such as HFD-fed rats [48], high-fat high-carbohydrate diet-fed rats [49], streptozotocin (STZ)-induced diabetes rats [50], and cholesterol-fed rabbits [51]. The lipid metabolism in hypercholesterolemic rats was improved by aqueous extract of ginger [52] and ginger powder [53]. Several studies reported the protective effects of bioactive compounds in ginger against dyslipidemia. 6-gingerol attenuated dyslipidemia in HFD-fed mice [54], obese db/db mice [55], and high-fat-diet fed rat [56]. In the HFD-fed golden hamster, 6-gingerol supplementation decreased the plasma TC, TG, and LDL-C levels, with concomitant inhibition of hepatic lipogenic genes [57]. Gingerol and shogaol-enriched extract decreased plasma TC and liver cholesterol via alteration of genes involved in cholesterol absorption and bile acid secretion in hamsters fed a high-cholesterol diet [58].
In the present study, SGE supplementation exerted hypoglycemic effects in DIO mice model. Several studies have reported the hypoglycemic properties of ginger and its bioactive compounds in animal models. Ethanolic extract of ginger attenuated hyperinsulinemia induced by liquid fructose in rats [59] and HFD-fed rats [48]. Furthermore, improved glucose and lipid profile by ethanolic extract of ginger in HFD-fed rats was associated with the alteration of hepatic genes involved in lipid metabolism [60]. Aqueous extract of ginger (500 mg/kg) exerted antihyperglycemic effects in STZ-induced diabetic rats [6162] and alloxan-induced diabetes rats [63]. Gingerol with HFD significantly decreased glucose, leptin, insulin, lipids levels in plasma, and liver lipids compared to control [56]. Wei et al. [37] compared the hypoglycemic effects of different active components of ginger, namely gingerols, shogaols, zingerone, and paradols. Among the active components, 6-paradol, the major metabolite of 6-shogaol, significantly reduced blood glucose and cholesterol in high-fat-diet-fed mice. These active compounds may responsible for the protective effect of SGE on increased serum lipid chemistry and hyperglycemia induced by a HFD.
Both adipocyte hypertrophy and hyperplasia contribute to adipose tissue expansion during the HFD challenge. In epididymal adipose tissue, hypertrophy is the prominent phenomenon contributing to adipose tissue expansion during the early stages of a HFD (within 1 month), and adipogenesis is apparent at the late stages of HFD [64]. In obese individuals and rodents, hypertrophy is closely related to the pathological expansion of adipose tissue. In the present study, SGE significantly reduced the weight and adipocyte size in epididymal adipose tissue than those of HFD. Similar to our results, ginger decreased adipocyte size and increased adipocyte number followed by suppressed macrophage infiltration and macrophage-associated cytokine in adipose fats of fructose-induced hyperinsulinemia in rats [59].
Differentiated adipocytes are post-mitotic; therefore, hyperplasia represents an increase in de novo adipocyte formation (adipogenesis). Adipocyte hypertrophy is closely linked to adipose dysfunction: this pathological expansion of white adipose tissue (WAT) is a significant component of the metabolic syndrome in obese individuals [264]. Therefore, we investigated whether the alterations in the expressions of genes involved in adipogenesis and TG synthesis are responsible for decreasing weight and adipocyte size in epididymal adipose tissue by SGE in DIO mice. The mRNA abundance of the adipogenic and lipogenic transcription factors and their downstream genes were significantly increased in epididymal adipose tissue in DIO mice. Interestingly, SGE supplementations attenuated the increases of epididymal gene expressions for adipogenesis and lipogenesis, i. e. PPARγ, C/EBPα, SREBP1-c, FAS, and ACC in DIO mice. Consistent with our findings, gingerenone A, a polyphenol in ginger, decreased the size of adipocyte in HFD-fed mice and reduced the expression of SREBP1 and FAS in epididymal adipose tissue [32]. The hot water extract of ginger ameliorated obesity and obesity-associated inflammation by lowering the mRNA levels of adipogenic genes and pro-inflammatory cytokines in white adipose tissue of rats fed a HFD [65]. Wang et al. [59] reported the ethanolic extract of ginger suppressed the expression of macrophage filtration marker but did not alter the adipogenic and lipogenic in adipose tissue of liquid fructose-fed rats. These results indicate extraction method may affect the different underlying mechanisms of the protective effect of ginger against obesity.
The lipolysis, the TG breakdown process, is essential in lipid metabolism and frequently altered with obesity [66]. In adipose tissue, the activity of lipases such as lipoprotein lipase (LPL), ATGL, and HSL are known to increase in obesity [6768]. Adipocyte lipolysis is a complex process mainly controlled by ATGL and HSL [68]. The mRNA abundance of ATGL and HSL in epididymal adipose tissue was significantly increased in SGE groups compared to the HFD group. Gingerenone A supplementation showed higher expression of ATGL and HSL in HFD-fed mice [32].
In the present study, the steamed ginger inhibited adipogenesis by regulating genes involved in adipocyte differentiation and lipogenesis in 3T3-L1 cells. Furthermore, steamed ginger exerted protective effects against adiposity in the epididymal adipose tissue of DIO mice by attenuating adipogenic and lipogenic genes. Further study is warranted to investigate which components of steamed ginger are responsible for these effects. In conclusion, the present study provides and supports that steamed ginger protects against high-fat DIO and metabolic disorders.

Notes

Funding: This work was supported by the Ministry of Agriculture, Food and Rural Affairs (MAFRA), through the 2015 Healthy Local Food Branding Project of the Rural Resources Complex Industrialization Support Program.

Conflict of Interest: The author declares no potential conflicts of interests.

Author Contributions:

  • Conceptualization: Cha YS, Kim B.

  • Data curation: Kim HJ, Kim B.

  • Formal analysis: Kim B, Kim HJ.

  • Funding acquisition: Cha YS.

  • Investigation: Kim B, Kim HJ.

  • Supervision: Cha YS.

  • Writing - original draft: Kim B, Kim HJ.

  • Writing - review & editing: Kim B, Cha YS.

References

1. Botchlett R, Woo SL, Liu M, Pei Y, Guo X, Li H, Wu C. Nutritional approaches for managing obesity-associated metabolic diseases. J Endocrinol. 2017; 233:R145–71. PMID: 28400405.
crossref
2. Jo J, Gavrilova O, Pack S, Jou W, Mullen S, Sumner AE, Cushman SW, Periwal V. Hypertrophy and/or hyperplasia: dynamics of adipose tissue growth. PLOS Comput Biol. 2009; 5:e1000324. PMID: 19325873.
crossref
3. Choe SS, Huh JY, Hwang IJ, Kim JI, Kim JB. Adipose tissue remodeling: its role in energy metabolism and metabolic disorders. Front Endocrinol (Lausanne). 2016; 7:30. PMID: 27148161.
crossref
4. Nishimura S, Manabe I, Nagasaki M, Hosoya Y, Yamashita H, Fujita H, Ohsugi M, Tobe K, Kadowaki T, Nagai R, Sugiura S. Adipogenesis in obesity requires close interplay between differentiating adipocytes, stromal cells, and blood vessels. Diabetes. 2007; 56:1517–1526. PMID: 17389330.
crossref
5. Haczeyni F, Bell-Anderson KS, Farrell GC. Causes and mechanisms of adipocyte enlargement and adipose expansion. Obes Rev. 2018; 19:406–420. PMID: 29243339.
crossref
6. Tandon P, Wafer R, Minchin JE. Adipose morphology and metabolic disease. J Exp Biol. 2018; 221(Suppl 1):221.
crossref
7. Roberts R, Hodson L, Dennis AL, Neville MJ, Humphreys SM, Harnden KE, Micklem KJ, Frayn KN. Markers of de novo lipogenesis in adipose tissue: associations with small adipocytes and insulin sensitivity in humans. Diabetologia. 2009; 52:882–890. PMID: 19252892.
8. Ali AT, Hochfeld WE, Myburgh R, Pepper MS. Adipocyte and adipogenesis. Eur J Cell Biol. 2013; 92:229–236. PMID: 23876739.
crossref
9. Mota de Sá P, Richard AJ, Hang H, Stephens JM. Transcriptional regulation of adipogenesis. Compr Physiol. 2017; 7:635–674. PMID: 28333384.
10. Siersbaek R, Nielsen R, Mandrup S. PPARgamma in adipocyte differentiation and metabolism--novel insights from genome-wide studies. FEBS Lett. 2010; 584:3242–3249. PMID: 20542036.
11. Musri MM, Párrizas M. Epigenetic regulation of adipogenesis. Curr Opin Clin Nutr Metab Care. 2012; 15:342–349. PMID: 22617562.
crossref
12. Liu Y, Sun M, Yao H, Liu Y, Gao R. Herbal medicine for the treatment of obesity: an overview of scientific evidence from 2007 to 2017. Evid Based Complement Alternat Med. 2017; 2017:8943059. PMID: 29234439.
crossref
13. Choi JY, Kim YJ, Cho SJ, Kwon EY, Ryu R, Choi MS. Metabolic effect of an oriental herbal medicine on obesity and its comorbidities with transcriptional responses in diet-induced obese mice. Int J Mol Sci. 2017; 18:18.
crossref
14. Ebrahimzadeh Attari V, Malek Mahdavi A, Javadivala Z, Mahluji S, Zununi Vahed S, Ostadrahimi A. A systematic review of the anti-obesity and weight lowering effect of ginger (Zingiber officinale Roscoe) and its mechanisms of action. Phytother Res. 2018; 32:577–585. PMID: 29193411.
crossref
15. Wang J, Ke W, Bao R, Hu X, Chen F. Beneficial effects of ginger Zingiber officinale Roscoe on obesity and metabolic syndrome: a review. Ann N Y Acad Sci. 2017; 1398:83–98. PMID: 28505392.
16. Ali BH, Blunden G, Tanira MO, Nemmar A. Some phytochemical, pharmacological and toxicological properties of ginger (Zingiber officinale Roscoe): a review of recent research. Food Chem Toxicol. 2008; 46:409–420. PMID: 17950516.
crossref
17. Lau AJ, Seo BH, Woo SO, Koh HL. High-performance liquid chromatographic method with quantitative comparisons of whole chromatograms of raw and steamed Panax notoginseng. J Chromatogr A. 2004; 1057:141–149. PMID: 15584233.
crossref
18. Toh DF, New LS, Koh HL, Chan EC. Ultra-high performance liquid chromatography/time-of-flight mass spectrometry (UHPLC/TOFMS) for time-dependent profiling of raw and steamed Panax notoginseng. J Pharm Biomed Anal. 2010; 52:43–50. PMID: 20079593.
crossref
19. Toh DF, Patel DN, Chan EC, Teo A, Neo SY, Koh HL. Anti-proliferative effects of raw and steamed extracts of Panax notoginseng and its ginsenoside constituents on human liver cancer cells. Chin Med. 2011; 6:4. PMID: 21255464.
crossref
20. Yoo KY, Lee CH, Li H, Park JH, Choi JH, Hwang IK, Kang IJ, Won MH. Ethyl acetate extracts of raw and steamed Codonopsis lanceolata protects against ischemic damage potentially by maintaining SOD1 and BDNF levels. Int J Neurosci. 2011; 121:503–509. PMID: 21671837.
21. Ho SC, Su MS. Evaluating the anti-neuroinflammatory capacity of raw and steamed garlic as well as five organosulfur compounds. Molecules. 2014; 19:17697–17714. PMID: 25365295.
crossref
22. Noda Y, Asada C, Sasaki C, Hashimoto S, Nakamura Y. Extraction method for increasing antioxidant activity of raw garlic using steam explosion. Biochem Eng J. 2013; 73:1–4.
crossref
23. Cheng XL, Liu Q, Peng YB, Qi LW, Li P. Steamed ginger (Zingiber officinale): changed chemical profile and increased anticancer potential. Food Chem. 2011; 129:1785–1792.
crossref
24. Iwasaki Y, Morita A, Iwasawa T, Kobata K, Sekiwa Y, Morimitsu Y, Kubota K, Watanabe T. A nonpungent component of steamed ginger--[10]-shogaol--increases adrenaline secretion via the activation of TRPV1. Nutr Neurosci. 2006; 9:169–178. PMID: 17176640.
crossref
25. Kim HJ, Kim B, Mun EG, Jeong SY, Cha YS. The antioxidant activity of steamed ginger and its protective effects on obesity induced by high-fat diet in C57BL/6J mice. Nutr Res Pract. 2018; 12:503–511. PMID: 30515278.
crossref
26. Park SH, Jung SJ, Choi EK, Ha KC, Baek HI, Park YK, Han KH, Jeong SY, Oh JH, Cha YS, Park BH, Chae SW. The effects of steamed ginger ethanolic extract on weight and body fat loss: a randomized, double-blind, placebo-controlled clinical trial. Food Sci Biotechnol. 2019; 29:265–273. PMID: 32064135.
crossref
27. Thomas SS, Kim M, Lee SJ, Cha YS. Antiobesity effects of purple perilla (Perilla frutescens var. acuta) on adipocyte differentiation and mice fed a high-fat diet. J Food Sci. 2018; 83:2384–2393. PMID: 30070698.
28. Kim M, Song SB, Cha YS. Effects of black adzuki bean (Vigna angularis, Geomguseul) extract on body composition and hypothalamic neuropeptide expression in rats fed a high-fat diet. Food Nutr Res. 2015; 59:27719. PMID: 26493717.
crossref
29. Kim M, Park JE, Song SB, Cha YS. Effects of black adzuki bean (Vigna angularis) extract on proliferation and differentiation of 3T3-L1 preadipocytes into mature adipocytes. Nutrients. 2015; 7:277–292. PMID: 25569623.
crossref
30. Campos CF, Duarte MS, Guimarães SE, Verardo LL, Wei S, Du M, Jiang Z, Bergen WG, Hausman GJ, Fernyhough-Culver M, Albrecht E, Dodson MV. Review: animal model and the current understanding of molecule dynamics of adipogenesis. Animal. 2016; 10:927–932. PMID: 26776309.
crossref
31. Li HX, Xiao L, Wang C, Gao JL, Zhai YG. Review: epigenetic regulation of adipocyte differentiation and adipogenesis. J Zhejiang Univ Sci B. 2010; 11:784–791. PMID: 20872986.
32. Suk S, Kwon GT, Lee E, Jang WJ, Yang H, Kim JH, Thimmegowda NR, Chung MY, Kwon JY, Yang S, Kim JK, Park JH, Lee KW. Gingerenone A, a polyphenol present in ginger, suppresses obesity and adipose tissue inflammation in high-fat diet-fed mice. Mol Nutr Food Res. 2017; 61:1700139.
crossref
33. Tzeng TF, Liu IM. 6-gingerol prevents adipogenesis and the accumulation of cytoplasmic lipid droplets in 3T3-L1 cells. Phytomedicine. 2013; 20:481–487. PMID: 23369342.
crossref
34. Tzeng TF, Chang CJ, Liu IM. 6-gingerol inhibits rosiglitazone-induced adipogenesis in 3T3-L1 adipocytes. Phytother Res. 2014; 28:187–192. PMID: 23519881.
crossref
35. Li C, Zhou L. Inhibitory effect 6-gingerol on adipogenesis through activation of the Wnt/β-catenin signaling pathway in 3T3-L1 adipocytes. Toxicol In Vitro. 2015; 30:394–401. PMID: 26498061.
crossref
36. Suk S, Seo SG, Yu JG, Yang H, Jeong E, Jang YJ, Yaghmoor SS, Ahmed Y, Yousef JM, Abualnaja KO, Al-Malki AL, Kumosani TA, Lee CY, Lee HJ, Lee KW. A bioactive constituent of ginger, 6-shogaol, prevents adipogenesis and stimulates lipolysis in 3T3-L1 adipocytes. J Food Biochem. 2016; 40:84–90.
crossref
37. Wei CK, Tsai YH, Korinek M, Hung PH, El-Shazly M, Cheng YB, Wu YC, Hsieh TJ, Chang FR. 6-paradol and 6-shogaol, the pungent compounds of ginger, promote glucose utilization in adipocytes and myotubes, and 6-paradol reduces blood glucose in high-fat diet-fed mice. Int J Mol Sci. 2017; 18:18.
crossref
38. Ahn EK, Oh JS. Inhibitory effect of galanolactone isolated from Zingiber officinale Roscoe extract on adipogenesis in 3T3-L1 cells. J Korean Soc Appl Biol Chem. 2012; 55:63–68.
crossref
39. Kuri-Harcuch W, Velez-delValle C, Vazquez-Sandoval A, Hernández-Mosqueira C, Fernandez-Sanchez V. A cellular perspective of adipogenesis transcriptional regulation. J Cell Physiol. 2019; 234:1111–1129. PMID: 30146705.
crossref
40. Ntambi JM, Young-Cheul K. Adipocyte differentiation and gene expression. J Nutr. 2000; 130:3122S–3126S. PMID: 11110885.
crossref
41. Farmer SR. Transcriptional control of adipocyte formation. Cell Metab. 2006; 4:263–273. PMID: 17011499.
crossref
42. Lee JE, Ge K. Transcriptional and epigenetic regulation of PPARγ expression during adipogenesis. Cell Biosci. 2014; 4:29. PMID: 24904744.
crossref
43. Kersten S. Mechanisms of nutritional and hormonal regulation of lipogenesis. EMBO Rep. 2001; 2:282–286. PMID: 11306547.
crossref
44. Semwal RB, Semwal DK, Combrinck S, Viljoen AM. Gingerols and shogaols: important nutraceutical principles from ginger. Phytochemistry. 2015; 117:554–568. PMID: 26228533.
crossref
45. Misawa K, Hashizume K, Yamamoto M, Minegishi Y, Hase T, Shimotoyodome A. Ginger extract prevents high-fat diet-induced obesity in mice via activation of the peroxisome proliferator-activated receptor δ pathway. J Nutr Biochem. 2015; 26:1058–1067. PMID: 26101135.
crossref
46. Fuhrman B, Rosenblat M, Hayek T, Coleman R, Aviram M. Ginger extract consumption reduces plasma cholesterol, inhibits LDL oxidation and attenuates development of atherosclerosis in atherosclerotic, apolipoprotein E-deficient mice. J Nutr. 2000; 130:1124–1131. PMID: 10801908.
crossref
47. Goyal RK, Kadnur SV. Beneficial effects of Zingiber officinale on goldthioglucose induced obesity. Fitoterapia. 2006; 77:160–163. PMID: 16513292.
crossref
48. Nammi S, Sreemantula S, Roufogalis BD. Protective effects of ethanolic extract of Zingiber officinale rhizome on the development of metabolic syndrome in high-fat diet-fed rats. Basic Clin Pharmacol Toxicol. 2009; 104:366–373. PMID: 19413656.
49. Li Y, Tran VH, Kota BP, Nammi S, Duke CC, Roufogalis BD. Preventative effect of Zingiber officinale on insulin resistance in a high-fat high-carbohydrate diet-fed rat model and its mechanism of action. Basic Clin Pharmacol Toxicol. 2014; 115:209–215. PMID: 24428842.
50. Bhandari U, Kanojia R, Pillai KK. Effect of ethanolic extract of Zingiber officinale on dyslipidaemia in diabetic rats. J Ethnopharmacol. 2005; 97:227–230. PMID: 15707757.
crossref
51. Bhandari U, Sharma JN, Zafar R. The protective action of ethanolic ginger (Zingiber officinale) extract in cholesterol fed rabbits. J Ethnopharmacol. 1998; 61:167–171. PMID: 9683348.
crossref
52. ElRokh SM, Yassin NA, El-Shenawy SM, Ibrahim BM. Antihypercholesterolaemic effect of ginger rhizome (Zingiber officinale) in rats. Inflammopharmacology. 2010; 18:309–315. PMID: 20730603.
crossref
53. Matsuda A, Wang Z, Takahashi S, Tokuda T, Miura N, Hasegawa J. Upregulation of mRNA of retinoid binding protein and fatty acid binding protein by cholesterol enriched-diet and effect of ginger on lipid metabolism. Life Sci. 2009; 84:903–907. PMID: 19379761.
crossref
54. Beattie JH, Nicol F, Gordon MJ, Reid MD, Cantlay L, Horgan GW, Kwun IS, Ahn JY, Ha TY. Ginger phytochemicals mitigate the obesogenic effects of a high-fat diet in mice: a proteomic and biomarker network analysis. Mol Nutr Food Res. 2011; 55(Suppl 2):S203–13. PMID: 21954187.
crossref
55. Son MJ, Miura Y, Yagasaki K. Mechanisms for antidiabetic effect of gingerol in cultured cells and obese diabetic model mice. Cytotechnology. 2015; 67:641–652. PMID: 24794903.
crossref
56. Saravanan G, Ponmurugan P, Deepa MA, Senthilkumar B. Anti-obesity action of gingerol: effect on lipid profile, insulin, leptin, amylase and lipase in male obese rats induced by a high-fat diet. J Sci Food Agric. 2014; 94:2972–2977. PMID: 24615565.
crossref
57. Tzeng TF, Liou SS, Chang CJ, Liu IM. [6]-gingerol dampens hepatic steatosis and inflammation in experimental nonalcoholic steatohepatitis. Phytomedicine. 2015; 22:452–461. PMID: 25925967.
crossref
58. Lei L, Liu Y, Wang X, Jiao R, Ma KY, Li YM, Wang L, Man SW, Sang S, Huang Y, Chen ZY. Plasma cholesterol-lowering activity of gingerol- and shogaol-enriched extract is mediated by increasing sterol excretion. J Agric Food Chem. 2014; 62:10515–10521. PMID: 25290252.
crossref
59. Wang J, Gao H, Ke D, Zuo G, Yang Y, Yamahara J, Li Y. Improvement of liquid fructose-induced adipose tissue insulin resistance by ginger treatment in rats is associated with suppression of adipose macrophage-related proinflammatory cytokines. Evid Based Complement Alternat Med. 2013; 2013:590376. PMID: 23533500.
crossref
60. de Las Heras N, Valero-Muñoz M, Martín-Fernández B, Ballesteros S, López-Farré A, Ruiz-Roso B, Lahera V. Molecular factors involved in the hypolipidemic- and insulin-sensitizing effects of a ginger (Zingiber officinale Roscoe) extract in rats fed a high-fat diet. Appl Physiol Nutr Metab. 2017; 42:209–215. PMID: 28125276.
61. Al-Amin ZM, Thomson M, Al-Qattan KK, Peltonen-Shalaby R, Ali M. Anti-diabetic and hypolipidaemic properties of ginger (Zingiber officinale) in streptozotocin-induced diabetic rats. Br J Nutr. 2006; 96:660–666. PMID: 17010224.
62. Abdulrazaq NB, Cho MM, Win NN, Zaman R, Rahman MT. Beneficial effects of ginger (Zingiber officinale) on carbohydrate metabolism in streptozotocin-induced diabetic rats. Br J Nutr. 2012; 108:1194–1201. PMID: 22152092.
63. Jafri SA, Abass S, Qasim M. Hypoglycemic effect of ginger (Zingiber officinale) in alloxan induced diabetic rats (Rattus norvagicus). Pak Vet J. 2011; 31:160–162.
64. Wang QA, Tao C, Gupta RK, Scherer PE. Tracking adipogenesis during white adipose tissue development, expansion and regeneration. Nat Med. 2013; 19:1338–1344. PMID: 23995282.
crossref
65. Kim S, Lee MS, Jung S, Son HY, Park S, Kang B, Kim SY, Kim IH, Kim CT, Kim Y. Ginger extract ameliorates obesity and inflammation via regulating microRNA-21/132 expression and AMPK activation in white adipose tissue. Nutrients. 2018; 10:10.
crossref
66. Frühbeck G, Méndez-Giménez L, Fernández-Formoso JA, Fernández S, Rodríguez A. Regulation of adipocyte lipolysis. Nutr Res Rev. 2014; 27:63–93. PMID: 24872083.
crossref
67. Wang H, Eckel RH. Lipoprotein lipase: from gene to obesity. Am J Physiol Endocrinol Metab. 2009; 297:E271–E288. PMID: 19318514.
crossref
68. Zechner R, Zimmermann R, Eichmann TO, Kohlwein SD, Haemmerle G, Lass A, Madeo F. FAT SIGNALS--lipases and lipolysis in lipid metabolism and signaling. Cell Metab. 2012; 15:279–291. PMID: 22405066.
TOOLS
Similar articles