+ All Categories
Home > Documents > Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic...

Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic...

Date post: 10-Mar-2021
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
11
Ginger prevents obesity through regulation of energy metabolism and activation of browning in high-fat diet-induced obese miceJing Wang a,b,c,d,e , Daotong Li a,b,c,d , Pan Wang a,b,c,d , Xiaosong Hu a,b,c,d,e , Fang Chen a,b,c,d, a College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China b National Engineering Research Center for Fruit and Vegetable Processing, China Agricultural University, Beijing, China c Key Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture, China Agricultural University, Beijing, China d Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, China e Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China Received 28 January 2019; received in revised form 8 April 2019; accepted 8 May 2019 Abstract Numerous natural herbs have been proven as safe anti-obesity resources. Ginger, one of the most widely consumed spices, has shown beneficial effects against obesity and related metabolic disorders. The present study aimed to examine whether the antiobesity effect of ginger is associated with energy metabolism. Mice were maintained on either a normal control diet or a high-fat diet (HFD) with or without 500 mg/kg (w/w) ginger supplementation. After 16 weeks, ginger supplementation alleviated the HFD-induced increases in body weight, fat accumulation, and levels of serum glucose, triglyceride and cholesterol. Indirect calorimetry showed that ginger administration significantly increased the respiratory exchange ratio (RER) and heat production in both diet models. Furthermore, ginger administration corrected the HFD-induced changes in concentrations of intermediates in glycolysis and the TCA cycle. Moreover, ginger enhanced brown adipose tissue function and activated white adipose tissue browning by altering the gene expression and protein levels of some brown and beige adipocyte-selective markers. Additionally, stimulation of the browning program by ginger may be partly regulated by the sirtuin-1 (SIRT1)/AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) pathway. Taken together, these results indicate that dietary ginger prevents body weight gain by remodeling whole-body energy metabolism and inducing browning of white adipose tissue (WAT). Thus, ginger is an edible plant that plays a role in the therapeutic treatment of obesity and related disorders. © 2019 Published by Elsevier Inc. Keywords: Ginger; Obesity; Energy metabolism; Browning; AMP-activated protein kinase (AMPK) 1. Introduction Obesity is increasing at an alarming rate and has become one of the greatest public health concerns all over the world. Estimates indicate that 38% of the world's adult population will be overweight in 2030 without effective intervention [1]. Obesity is caused by an imbalance of energy intake and expenditure, and thus, the best way to treat obesity is to cause a decrease in the former and an increase in the latter [2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose tissue are known-white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue. WAT is optimized to store energy in the form of large droplets of triglycerides (TGs) for later use and regulate the whole-body homeostasis [3]. BAT contains a high density of mitochondria with uncoupling protein-1 (UCP1) that uncouples mitochondrial oxidative respiration from ATP production to produce heat [4,5]. Original white adipocytes can be transformed into beige adipocytes (browning process), which have similar characteristics to brown adipocytes, such as higher respiratory rate, thermogenesis rate and energy consumption rate [6]. Recent studies indicated that browning of WAT is regulated by several genes, such as UCP1, PR domain containing 16 (PRDM16), cell death-inducing DFFA-like effector a (CIDEA) and PGC-1α. The expression of PGC-1α is regulated by AMPK and SIRT1, which are two important nutrient and energy sensors [7]. Thus, SIRT1, AMPK and PGC-1α form an energy sensing network that controls energy expenditure in mitochondria [8]. Thus, promoting adipose tissue browning may offer a viable approach to counteracting obesity. Currently, several phytochemicals and plant-derived foods, such as capsaicin [9], curcumin [10], cranberry [11], and rose [12], are known to affect energy homeostasis by browning of WAT and reduce the risk of overweight and obesity. Ginger, derived from the rhizome of Available online at www.sciencedirect.com ScienceDirect Journal of Nutritional Biochemistry 70 (2019) 105 115 Conict of interest statement: All authors have declared no conicts of interest. Corresponding author at: College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Haidian District, Beijing 100083, China. Tel./fax: +86 10 62737645 18. E-mail address: [email protected] (F. Chen). https://doi.org/10.1016/j.jnutbio.2019.05.001 0955-2863/© 2019 Published by Elsevier Inc.
Transcript
Page 1: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Available online at www.sciencedirect.com

ScienceDirect

istry 70 (2019) 105–115

Journal of Nutritional Biochem

Ginger prevents obesity through regulation of energy metabolism and activation ofbrowning in high-fat diet-induced obese mice☆

Jing Wanga,b, c,d,e, Daotong Lia,b, c,d, Pan Wanga,b, c,d, Xiaosong Hua,b, c,d, e, Fang Chena,b, c,d,⁎aCollege of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China

bNational Engineering Research Center for Fruit and Vegetable Processing, China Agricultural University, Beijing, ChinacKey Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture, China Agricultural University, Beijing, China

dEngineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, ChinaeBeijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China

Received 28 January 2019; received in revised form 8 April 2019; accepted 8 May 2019

Abstract

Numerous natural herbs have been proven as safe anti-obesity resources. Ginger, one of the most widely consumed spices, has shown beneficial effectsagainst obesity and related metabolic disorders. The present study aimed to examine whether the antiobesity effect of ginger is associated with energymetabolism. Mice were maintained on either a normal control diet or a high-fat diet (HFD) with or without 500 mg/kg (w/w) ginger supplementation. After 16weeks, ginger supplementation alleviated the HFD-induced increases in body weight, fat accumulation, and levels of serum glucose, triglyceride and cholesterol.Indirect calorimetry showed that ginger administration significantly increased the respiratory exchange ratio (RER) and heat production in both diet models.Furthermore, ginger administration corrected the HFD-induced changes in concentrations of intermediates in glycolysis and the TCA cycle. Moreover, gingerenhanced brown adipose tissue function and activated white adipose tissue browning by altering the gene expression and protein levels of some brown andbeige adipocyte-selective markers. Additionally, stimulation of the browning program by ginger may be partly regulated by the sirtuin-1 (SIRT1)/AMP-activatedprotein kinase (AMPK)/peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) pathway. Taken together, these results indicate that dietary gingerprevents body weight gain by remodeling whole-body energy metabolism and inducing browning of white adipose tissue (WAT). Thus, ginger is an edible plantthat plays a role in the therapeutic treatment of obesity and related disorders.© 2019 Published by Elsevier Inc.

Keywords: Ginger; Obesity; Energy metabolism; Browning; AMP-activated protein kinase (AMPK)

1. Introduction

Obesity is increasing at an alarming rate and has become one of thegreatest public health concerns all over the world. Estimates indicatethat 38% of the world's adult population will be overweight in 2030without effective intervention [1]. Obesity is caused by an imbalanceof energy intake and expenditure, and thus, the best way to treatobesity is to cause a decrease in the former and an increase in the latter[2]. Adipose tissue is a major metabolic tissue and plays a key role inenergy homeostasis. Currently, three types of adipose tissue areknown-white adipose tissue (WAT), brown adipose tissue (BAT), andbeige adipose tissue. WAT is optimized to store energy in the form oflarge droplets of triglycerides (TGs) for later use and regulate thewhole-body homeostasis [3]. BAT contains a high density ofmitochondria with uncoupling protein-1 (UCP1) that uncouplesmitochondrial oxidative respiration from ATP production to produce

☆ Conflict of interest statement: All authors have declared no conflicts of intere⁎ Corresponding author at: College of Food Science andNutritional Engineering, Ch

100083, China. Tel./fax: +86 10 62737645 18.E-mail address: [email protected] (F. Chen).

https://doi.org/10.1016/j.jnutbio.2019.05.0010955-2863/© 2019 Published by Elsevier Inc.

heat [4,5]. Original white adipocytes can be transformed into beigeadipocytes (browning process), which have similar characteristics tobrown adipocytes, such as higher respiratory rate, thermogenesis rateand energy consumption rate [6]. Recent studies indicated thatbrowning of WAT is regulated by several genes, such as UCP1, PRdomain containing 16 (PRDM16), cell death-inducing DFFA-likeeffector a (CIDEA) and PGC-1α. The expression of PGC-1α is regulatedby AMPK and SIRT1, which are two important nutrient and energysensors [7]. Thus, SIRT1, AMPK and PGC-1α form an energy sensingnetwork that controls energy expenditure in mitochondria [8]. Thus,promoting adipose tissue browning may offer a viable approach tocounteracting obesity.

Currently, several phytochemicals and plant-derived foods, such ascapsaicin [9], curcumin [10], cranberry [11], and rose [12], are knownto affect energy homeostasis by browning of WAT and reduce the riskof overweight and obesity. Ginger, derived from the rhizome of

st.ina Agricultural University, No. 17, Qinghua East Road, Haidian District, Beijing

Page 2: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Table 1The chemical and nutritional profile of ginger

Components Concentration(mg/g)

Moisture 68.71±0.86Ash 122.13±4.72Fat 57.91±2.62Dietary fiber 154.82±4.42Protein 150.06±9.24Total sugar 69.85±1.74Total phenolics (mg GAE/g d.w.) 14.27±0.28Total flavonoids (mg Rutin/g d.w.) 15.49±0.266-Gingerol 7.03±0.018-Gingerol 2.21±0.0110-Gingerol 3.89±0.026- Shogaol 0.87±0.01

106 J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

Zingiber officinaleRosco (Zingiberaceae), is awell-known spice that hasbeen widely used for culinary purposes [13]. It contains variousphytochemicals and biologically active components, such as gingerolsand shogaols [14]. Apart from other traditional medical uses, gingerand itsmain bioactive constituents were reported to attenuate obesityboth in rodent animal models and cell lines [15]. For example, gingerextract administration can prevent HFD-induced obesity [16] andfructose overconsumption-induced insulin resistance in rats [17] byregulating peroxisome proliferator-activated receptors (PPARs) andthe inflammatory signaling pathway in adipocytes. Moreover, 6-gingerol and 6-shogaol, two major bioactive constituents of ginger,also exhibited antiobesity effects by altering the activities of some lipidmetabolismmarker enzymes and decreasing the expression of variouslipogenicmarker proteins in both rats and cell lines [18–21]. However,whether ginger supplementation can influence WAT browning inobese mice has never been explored.

In the present study, we analyzed the effects of ginger on energymetabolites involved in glycolysis and the TCA cycle and investigatedthe changes in some key gene and protein expression levels followingginger treatment. Finally, we proposed the molecular mechanism ofthe ginger-induced browning effect. The present study could providenew insight into the antiobesity function of ginger.

2. Materials and methods

2.1. Ethics statement

All animal experimental procedures followed in this study werereviewed and approved by the Biomedical Ethical Committee ofPeking University (Beijing, China) with an approval number(LA2018288).

2.2. Preparation and analysis of chemical components of ginger

Fresh ginger rhizomes (Zingiber officinale Rosco) were purchasedfrom a local market in Laiwu, Shandong Province, China, in February2018. Ginger samples were authenticated by Prof. Fang Chen based onthe monograph of ginger documented in the Chinese Pharmacopeia(2015 edition). A total of 2 kg of fresh ginger rhizomes were peeled,washed, further cut into pieces and then dried with a freeze-dryer(LGJ-10C, Beijing, China) for 48 h. The dried samples were subse-quently milled using a commercial hand-carry milling machine toproduce ginger powder, which was then stored in a moisture-controlled cabinet.

The protein, lipid, water and total sugar content of ginger weredetermined by the Kjeldahlmethod, the Soxhlet extractormethod, theambient pressure drying method and the dinitrosalicylic acid (DNS)colorimetricmethod, respectively. The ash contentwasmeasured by acombination of ashing and gravimetric procedures. The dietary fibercontent of ginger was determined by using a combination ofenzymatic and gravimetric procedures according to AOAC. The totalphenolic and flavonoid contents were determined by using the Folin–Ciocalteu method and the aluminum chloride colorimetric method,respectively (Table 1).

2.3. Identification and quantification of the main phenolic compounds inginger by reversed-phase high-performance liquid chromatography(HPLC)

HPLC was performed to identify and quantitate the majorcompounds in ginger using an RF-10AXL HPLC system (ShimadzuCo., Japan). Chromatographic separations were performed on areversed-phase column (Sunfire™ C18, 250×4.6 mm ID, 5 μm). The

mobile phases consisted of 2 phases: water (A) and acetonitrile (B).The gradient program forHPLCwas as follows: 0–5min, 0–20%B; 5–45min, 20–90% B; and 45–70 min, 100% B. The injection volume was 20μL with a 1 mL/min flow. The column temperature was maintained at30°C, and the detection wavelength was 280 nm. The chromatogramsof the major compounds from ginger are presented in SupplementaryFig. S1, and the main phenolic compounds concentrations are shownin Table 1. The 6-, 8-, and 10-gingerols and 6-shogaol standards(purityN98%) were purchased from Chromadex (Irvine, CA, USA).Unless noted, all chemicals were purchased from Sigma-Aldrich (St.Louis, MO, USA).

2.4. Animals and experimental design

Five-week-old C57BL/6 Jmalemice (provided by PekingUniversityAnimal Breeding Unit) were housed under standard laboratoryconditions (25±2°C, 12-h light/dark cycle) with free access to foodand drinking water. After acclimatization for one week, animals wererandomly divided into four dietary groups (n=8 per group). Twogroups were fed with a normal control diet (NCD; 10% kcal from fat,D12450B; Research Diets) or an identical diet supplemented withginger (NCD-G). The other two groups were fed a HFD (60% kcal fromfat, D12492; Research Diets) or an identical diet supplemented withginger (HFD-G). The ingredients and energy densities of theexperimental diets are shown in Supplementary Table S2. In thisstudy, we investigated the effects of ginger onWAT browning in obesemice with the dose of 500mg/kg according to the previous study [22].In each feeding condition, treatment groups were orally administered500 mg/kg body weight ginger once daily, whereas the other twogroups were used as controls and gavaged with an equal volume ofnormal saline. Body weight and food intake were assessed once perweek. After 16 weeks, mice were euthanized by carbon dioxideanesthesia followed by cervical dislocation. Blood was drawn andimmediately centrifuged (3000 rpm, 10 min). Epididymal whiteadipose tissue (Epi-WAT), perirenal white adipose tissue (Per-WAT),inguinal subcutaneous adipose tissue (Ing-SAT) and BAT were rapidlyisolated, weighed and stored at−80°C until further use.

2.5. Biochemical analysis

For biochemical analyses, the blood samples were centrifuged at3000 rpm for 10 min at 4°C. The supernatant was stored at −80°C. A3100 automatic biochemistry analyzer (Hitachi Ltd., Tokyo, Japan)wasused to determine serum concentrations of glucose (GLU), totalcholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL),high-density lipoprotein (HDL), alanine aminotransferase (ALT), andaspartate aminotransferase (AST).

Page 3: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

107J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

2.6. Histology and immunofluorescence staining of tissue sectionsanalysis

After mice were sacrificed, BAT and Epi-WAT were collected andfixed in 4% paraformaldehyde; then the tissues were paraffin-embedded and sectioned at 5–7 μm. Hematoxylin and eosin (H&E)staining was performed according to a standard method. Forimmunofluorescence, briefly, BAT and Epi-WAT sections wereincubated with UCP1 (Sigma), diluted at 1:100, at 4°C for 12 h, andthen incubated with a secondary antibody for 1 h. Stained slides wereviewedunder a Zeiss Observer.A1Microscope (Carl Zeiss, Oberkochen,Germany) at ×200magnification, and an Olympus C4000 zoomdigitalcamera (Nikon DS-L1, Nikon, Tokyo, Japan) was used to capture theimages.

2.7. Metabolic phenotyping

Two days before tissue sample collection, mice were placed inmetabolic chambers (Columbus Instruments, Columbus, OH) andacclimated over a 24 h period. Then, the metabolic rate, includingoxygen consumption (VO2) and carbon dioxide production (VCO2),was monitored every 13 min for the duration of the next 24 h. Therespiratory exchange ratio (RER) was calculated as the ratio of carbondioxide volume versus oxygen volume (VCO2/VO2). In addition, heatand spontaneous locomotor activity were closely monitored for theduration of the measurement period.

2.8. GC–MS analysis of target TCA metabolites in serum

A total of 20 μL of thawed serum and 80 μL of cold methanolincluding an internal standard (10 μg/mL dulcitol)were combined andvortexed for 60 s. After centrifugation at 14,000 × g and 4°C for 15min, 70 μL of supernatant was evaporated to dryness under a nitrogenstream. The residue was reconstituted in 30 μL of 20 mg/mLmethoxyamine hydrochloride in pyridine, and the resulting mixturewas incubated at 37°C for 90 min. Next, 30 μL of N,O-bis(trimethylsi-lyl)trifluoroacetamide (BSTFA) (with 1% trimethylchlorosilane(TMCS)) was added to the mixture and derivatized at 70°C for 60min prior to GC–MS analysis.

A mixed standard solution, containing 20 μg/mL succinic acid,fumaric acid, malic acid, citric acid, cis-aconitic acid, 2-ketoglutaricacid, pyruvic acid, lactic acid, glutamic acid, glutamine, pantothenicacid, ribose-5-phosphate, glucose, glucose-6-phosphate and fructose-6-phosphate (Sigma-Aldrich), was prepared in water and seriallydiluted to 0.02–10 μg/mL. Then, 70 μL of the standardmixture, 35 μL ofthe quality control (QC) sample and 28 μL of 10 μg/mL dulcitol weremixed and evaporated to dryness under a nitrogen stream. The othersteps were the same as those for serum.

An instrumental analysis was performed on an Agilent 7890A gaschromatography system coupled to an Agilent 5975C inert MSDsystem (Agilent Technologies, CA, USA). An OPTIMA® 5 MS Accentfused-silica capillary column (30 m×0.25 mm×0.25 μm; MACHEREY-NAGEL, Düren, GER) was utilized to separate the derivatives. Helium(N 99.999%) was used as a carrier gas at a constant flow rate of 1 mL/min through the column. Injection volume was 1 μL in a split ratio of10:1 for glucose and lactic acid and 1 μL in splitless mode for the othermetabolites. The solvent delay time was 5.5 min. The initial oventemperature was held at 60°C for 1 min, ramped to 240°C at a rate of12°C/min, to 320°C at 40°C/min and finally held for 4 min. Thetemperatures of the injector, transfer line, and electron impact ionsource were set to 250°C, 260°C, and 230°C, respectively. The impactenergy was 70 eV, and data were collected in both full-scan (m/z50–600) and SIM mode. Energy metabolites were analyzed by MSDChemStation Version E.02.02.1431 (Agilent Technologies). The quan-titation ions for all target compounds, including the internal standard,

are shown in Supplementary Table S3, which also includes thechromatographic retention times and relative standard deviation (%RSD).

2.9. Quantitative real-time polymerase chain reaction (PCR) analysis

Total RNAwas extracted fromdifferent adipose tissues using TRIzolreagent (Invitrogen, Carlsbad, USA) according to the manufacturer'sinstructions. The quantification and qualitative ratiometric analysis ofRNA was performed using a Nanodrop 2000 (Thermo Scientific,Wilmington, DE, USA). For each sample, 1 μg of total RNAwas reverse-transcribed with a High-Capacity cDNA Reverse Transcription kit(Tiangen Biotech Co., Ltd). Gene expression was quantitated using aFast Start Essential DNA Green Light Master kit (Roche, Indianapolis,IN, USA) in a Light Cycler 480 real-time PCR system (Roche). The PCRconditionswere as follows: 10min at 95°C, followed by 40 cycles of 10s at 95°C, annealing for 10 s at 60°C and extension for 10 s at 72°C. Dataanalysis was conducted by the 2-ΔΔCT method. β-Actin was used asthe reference gene for normalization. The sequences of the sense andantisense primers used for amplification are shown in SupplementaryTable S4.

2.10. Western blot analysis

Tissues were incubated in RIPA lysis buffer (Thermo Fisher,Waltham, MA, USA) and phosphatase inhibitor (Sigma-Aldrich, SaintLouis, MN, USA) at pH 7.4 for 30 min. The samples were subsequentlysonicated at 4°C. After centrifuging the samples (12,000×g for 15 minat 4°C), the protein concentrations of the supernatant were measuredusing a BCA protein assay kit (Pierce, Rockford, IL, USA) according tothe manufacturer's protocol. Equal amounts of protein (40 μg) fromeach sample were loaded and separated by SDS-PAGE and transferredonto a PVDF membrane using Bio-RAD electrophoresis equipment.Membranes were blocked with 5% nonfat milk in Tris-buffered salineand Tween 20 (TBST) for 1 h at room temperature. Themembranewasthen rinsed three times consecutively with TBST buffer, followed byincubation for 1 h with 1:1000 dilutions of primary monoclonalantibodies: anti-UCP1 (Abcam), anti-β-actin, anti-p-AMPK, anti-AMPK, anti-SIRT1, anti-PRDM16 and anti-PGC-1α (Cell SignalingTechnology)-in TBST buffer containing 1% skim milk. After threewashes, the membrane was incubated for 1 h with horseradishperoxidase-conjugated anti-rabbit IgG secondary antibody (1:2000,Cell Signaling Technology) in TBST buffer containing 1% skimmilk. Allsignals were visualized and analyzed by densitometric scanning(Image Quant TL7.0, GE Healthcare Bio-Sciences AB). The density ofthe bandswas quantified using ImageJ Software (National Institutes ofHealth, USA). The ratio of the intensity of the target protein to that ofthe β-actin loading control was calculated to represent the expressionlevel of the protein.

2.11. Statistical analysis

All data are expressed as themean±standard error of themean (S.E.M.). Statistical analyses were conducted using SPSS software (IBMCorporation, NY, USA) version 20. Differences between groups wereanalyzedwith one-way analysis of variance (ANOVA), followed by theleast significant difference (LSD) post hoc test. P values less than .05were considered indicative of significance.

3. Results

3.1. Effect of ginger on obesity-induced metabolic disorders in mice

After the 16-week feeding period, ginger supplementation showeda protective role against the development of obesity in mice (Table 2).

Page 4: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Table 2Effect of ginger administration on obesity-related parameters.

Variables/Groups NCD NCD-G HFD HFD-G

Final body weight (g) 27.98±0.33a 26.00±0.41a 36.39±0.66c 32.43±0.60b

Weight gain (g) 9.25±0.28a 7.44±0.39a 17.04±0.83c 13.90±0.43b

Total energy intake(Kcal)

9 4 4 . 5 0 ±14.50a

9 4 8 . 1 6 ±7.00a

1 1 8 8 . 7 7±5.35b

1 1 7 9 . 5 0±9.48b

Energy efficiency (g/Kcal)

0 . 0 2 9 6 ±0.0003b

0 . 0 2 7 4 ±0.0004a

0 . 0 3 0 6 ±0.0005b

0 . 0 2 7 4 ±0.0005a

Epi-WAT/body weight(mg/g)

17.57±1.11a 16.18±0.74a 54.48±2.41c 31.87±2.00b

Per-WAT/body weight(mg/g)

4.91±0.69a 4.47±0.52a 24.44±2.37c 11.85±0.88b

Ing-SAT/body weight(mg/g)

9.10±1.31a 7.22±1.01a 26.14±1.80b 10.83±1.53a

All data are expressed as the mean±S.E.M. (n=8). Statistical analysis was performedusing ANOVA. The means with different superscripts are considered significantlydifferent (Pb.05)

Table 3Serum biochemical analyses fromNCD and HFDmice with or without ginger treatment.

NCD NCD-G HFD HFD-G

Fasting glucose (mmol/L)

4.98±0.42b 2.48±0.13a 7.69±0.82c 5.56±0.43b

Insulin (ng/mL) 0.87±0.01b 0.82±0.01a 1.10±0.02c 0.90±0.00b

HOMA-IR 4.08±0.34a 1.91±0.11a 7.89±0.81c 4.70±0.37b

TC(mmol/L) 3.41±0.12a 3.27±0.08a 4.57±0.18c 3.91±0.18b

TG(mmol/L) 0.33±0.05b 0.14±0.02a 0.68±0.06c 0.27±0.02a

HDL-C(mmol/L) 2.72±0.10ab 2.59±0.10a 3.52±0.14c 2.93±0.13b

LDL-C(mmol/L) 0.43±0.06a 0.58±0.05a 0.91±0.05b 1.00±0.10b

ALT(U/L) 3 2 . 8 8 ±5.22bc

18.88±1.22a 40.25±2.28c 30.25±2.17b

AST(U/L) 1 1 9 . 8 8 ±6.40b

1 1 4 . 7 5 ±5.47ab

1 3 1 . 6 3 ±8.08c

1 0 5 . 5 0 ±2.24a

Values are expressed as the mean±S.E.M. (n=8). Statistical analysis was performedusing ANOVA. The means with different superscripts are considered significantlydifferent (Pb.05).

108 J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

First, no significant difference in body weight was found between theNCD-G group and NCD group. However, the final body weight, bodyweight gain and energy efficiency tended to be lower in the HFD-Ggroup than in the HFD group.We then isolated the threemain types ofwhite adipose tissue frommice: Epi-WAT, Per-WAT, and Ing-SAT (Fig.1). Consistent with the body weight gain, weights of all three types ofadipose tissues and the ratio of various adipose tissues normalized tobody weight were clearly increased by HFD and decreased by gingersupplementation in mice, indicating a protective role of ginger in thedevelopment of obesity. Moreover, ginger was highly effective inrecovering theHFD-induced increase in levels of serumglucose, TC, TGand HDL-C as well as liver AST and ALT levels (Table 3).

3.2. Effect of dietary ginger on energy expenditure in mice

The metabolic rates of mice were further analyzed by open-circuitindirect calorimetry. Compared with the NCD group, the HFD grouppresented lower O2 consumption and CO2 production. However,ginger supplementation significantly elevated themetabolic rate bothin NCD and HFD mice (Fig. 2A, B). Next, RER was used to estimate therelative contribution of fat and carbohydrates to whole-body energymetabolism in mice. As expected, mice treated with ginger revealed asmall increase during the nocturnal activity phase in both diet models(Fig. 2C), and heat production was also increased (Fig. 2D). Consistent

Fig. 1. Representative image

with the elevation in metabolic rates, ginger supplementationsignificantly increased the ambulatory activity of the animals duringthe night phase (Fig. 2E). These data indicate that ginger attenuatedHFD-induced obesity due to enhanced energy expenditure.

3.3. Effect of dietary ginger on serum metabolites involved in glycolysisand the TCA cycle in mice

In this study, a novel, specific and sensitive GC–MS assay formetabolites involved in glycolysis and the TCA cycle was developed.The GC–MS chromatograms of the 13 target compounds in thestandard solutions and a representative serum sample are shown inSupplementary Fig. S5. The concentrations of the major intermediatesinvolved in glycolysis, such as fructose-6-phosphate (F6P), glucose-6-phosphate (G6P) and ribose-5-phosphate (R5P), were increased inHFD rats compared with NCD rats, and ginger administrationcorrected the changes in glycolysis induced by a HFD. In addition,HFD-fed mice also displayed reduced concentrations of TCA cycleintermediates in serum, such as pyruvic acid, lactic acid, succinic acid,glutamate, glutamic acid, fumaric acid and malic acid. Clearly,treatment with ginger reversed the levels of intermediates (Fig. 3),suggesting that ginger positively modulated the TCA cycle. Consider-ing the above results, we speculated that ginger improved metabolicimpairments via the glycolysis/gluconeogenesis-TCA cycle.

s of WATs and BAT.

Page 5: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Fig. 2. Changes inwhole-body energy balance in response to ginger supplementation. Indirect calorimetrymeasures of 24-h (A) oxygen consumption, (B) carbon dioxide production, (C)RER, (D) heat production and (E) voluntary activity. The results are expressed as themeans±S.E.M. (n=3). Differenceswere assessed byANOVA anddenoted as follows:&Pb.05,&&Pb.01,&&&Pb.001, HFD-G vs HFD; #Pb.05, ##Pb.01, ###Pb.001, HFD vs. NCD; ⁎Pb.05,⁎⁎Pb.01, ⁎⁎⁎Pb.001, NS not significant, NCD-G vs NCD.

109J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

3.4. Ginger can promote browning of WAT and activate BAT in mice

Further, we determined whether dietary ginger stimulated thegeneration of brown-like adipocytes. The results showed that gingersupplementation upregulated the expression of a series of brownadipocyte-specific genes (UCP1, PRDM16 and CIDEA) and beigeadipocyte-selective marker genes (TMEM26 and CITED1) in bothWAT and BAT of obese mice. Moreover, ginger increased the mRNAexpression of PGC-1α, the master regulator of mitochondrialbiogenesis and oxidative phosphorylation. Consistently, ginger pro-moted the expression of genes involved in mitochondrial biogenesis,such as TFAM and NRF1 (Fig. 4A, B).

The immunofluorescence staining results showed that the expres-sion of UCP1 in the ginger-treated groups was higher than that in thecorresponding control groups (Fig. 4G). This finding was in accordwith the Western blotting results demonstrating that the UCP1protein level was increased in WAT and BAT after ginger supplemen-tation (Fig. 4C-F). Moreover, the protein expression of PRDM16 andPGC-1αwas also markedly increased in both WAT and BAT of ginger-treated mice (Fig. 4C-F). These data indicated that ginger could

increase the BAT thermogenic progress and induce adipocytebrowning in WAT. Additionally, HFD decreased AMPK phosphoryla-tion without an effect on total AMPK levels, resulting in a decrease inthe p-AMPK/AMPK ratio, which was recovered by ginger. A similarmodulation of SIRT1 was observed after ginger treatment (Fig. 4E, F).Together, these results provide evidence that administration of gingerinduced browning in WAT of mice by activating AMPK.

4. Discussion

Browning of WAT can increase energy expenditure through UCP1-mediated thermogenesis. Thus, pharmacologic or nutritional strate-gies for promoting adipocyte browning and thermogenesis mayprovide a defense against obesity and associated metabolic diseases[23]. Growing evidence indicates that ginger exerts beneficial effectsagainst obesity and relatedmetabolic syndromes [15,16,24], but only asmall number of studies focus on energy metabolism. Dietary gingerextract was previously shown to increase energy expenditure andattenuate diet-induced obesity in C57BL/6 Jmice through activation ofthe PPAR-δ pathway [16]. Moreover, an in vitro mechanistic study

Page 6: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Fig. 3. Potential metabolic pathways disturbed in HFD-induced obesemice and alterations by ginger treatment. The results are expressed as themeans±S.E.M. (n=8). Differences wereassessed by ANOVA and denoted as follows: &Pb.05, &&Pb.01, &&&Pb.001, HFD-G vs HFD; #Pb.05, ##Pb.01, ###Pb.001, HFD vs. NCD; ⁎Pb.05,⁎⁎Pb.01, ⁎⁎⁎Pb.001, NS not significant, NCD-G vsNCD. No mark = no significant difference.

110 J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

showed that 6-gingerol dose-dependently increased AMPKα-subunitphosphorylation accompanied by a time-dependent marked increasein PGC-1αmRNA expression and mitochondrial content in L6 skeletalmuscle cells [25]. Nevertheless, whether ginger supplementation caninfluenceWAT browning in obese mice and if so, what the underlyingmechanisms are remain unclear.

The present study demonstrated that ginger prevented obesity byenhancing energy expenditure and activating browning and thermo-genic functions. First, ginger noticeably lowered HFD-induced in-creases in body weight gain, fat accumulation, and serum glucose, TC,TG and LDL-C levels. Consistently, ginger supplementation signifi-cantly alleviated the HFD-induced increase in the size of adipocytes. Inaddition, ginger treatment dramatically increased RER and heatproduction.

Next, we evaluated the impact of ginger supplementation onenergy metabolism. Recently, many nontargeted metabolomicsanalyses have suggested that obesity is associated with the levels ofmetabolic intermediates in energy metabolism pathways, includingglycolysis and the TCA cycle [26,27]. Metabolites such as pyruvate,lactate, and citratewere themain discriminatingmetabolites betweenthe obese and the healthy models [28]. Therefore, the present studyshowed that HFD clearly distorted glucose metabolism by increasingthe concentrations of the major intermediates involved in thephosphogluconate pathway. Moreover, certain other key metabolitesrelated to the TCA cycle were reduced in obese rats. These findings areconsistent with previous results obtained in nontargeted metabolo-mics studies of obesity [26,29]. Interestingly, ginger supplementationsignificantly lowered the concentrations of the metabolic intermedi-ates involved in glucose metabolism, such as R5P, G6P and F6P. Theseresults demonstrated that ginger could improve distorted glucosemetabolism, which was further supported by the reduction in serumglucose levels. Moreover, dietary ginger could greatly attenuatemetabolic impairments by ameliorating the blood lipid profile and

reducing lipid accumulation, as well as elevating concentrations ofsome metabolites involved in the TCA cycle, such as pyruvate andlactate. Among these metabolites, pyruvate plays a key role in glucoseaerobic oxidation and energy production [30], as it not only is a keymetabolite in glucose metabolism but also can serve as an importantsource of acetyl-CoA to fuel the TCA cycle. As the product of pyruvate,lactate in particular was increased after ginger treatment of obesemice. Notably, lactate can induce UCP1 gene expression in whiteadipocytes and the expression of genes associated with fatty acidoxidation andmitochondrial function, indicative of the recruitment ofa thermogenic profile [31]. Consistently, Morinda citrifolia L. leafextract, commonly called noni or Indian mulberry, improved HFD-induced perturbations in various metabolic pathways, predominantlyin the glucose and TCA cycle, as reflected by positive modulation oflactate, pyruvate, and glucose levels [32]. Another important inter-mediate in the TCA cycle is citric acid, which is regulated by insulin,glucose level, fatty acid utilization, and cholesterol synthesis [33]. Theincreased level of citric acid in obese mice supplemented with gingerfurther indicated a restoration of the glycolytic and energy metabolicdysfunction induced by HFD [34]. Alterations in some other metab-olites (such as 2-ketoglutaric acid, succinic acid, fumaric acid,glutamine and malic acid levels) caused by HFD-induced obesity incertain metabolic pathways were also notably improved upontreatment with ginger. Therefore, ginger could prevent energymetabolism impairment caused by HFD.

The TCA cycle is coupled with ATP production and proton leak,which can release energy in the form of heat [35]. UCP1, one of themain mitochondrial proteins, can dissipate the proton gradientgenerated by the NADH-powered pumping of protons from themitochondrial matrix to the mitochondrial intermembrane space andlet H+directly pass through the inner-membrane ofmitochondria andrelease heat [36]. Furthermore, BAT thermogenesis is dependent onUCP1 since it confers to brown adipocytes the specific capacity to

Page 7: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

111J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

dissipate oxidation energy as heat [37]. Improvement of thermogeniccapacity through activation of BAT may be a highly beneficialtherapeutic strategy for counteracting obesity and its complications[38,39]. Many natural compounds and food ingredients, such asresveratrol, cinnamaldehyde, sesamol and spirulina maxima extract,have shown antiobesity effects through activation of browning andincreases in energy expenditure [39–42]. Evidences from the presentstudy indicated that the beneficial effects of ginger on suppression ofobesity-related pathological factors were associated with BAT activa-tion. The expression of UCP1 was increased in BAT and emerged inWAT after ginger treatment, which was further confirmed by UCP1immunofluorescence staining. Moreover, ginger administration up-regulated several other dominant transcriptional regulators of

Fig. 4. Dietary ginger supplementation induced brown fat-like changes inWAT and BAT of mice(B) (n=6); Protein level of brown adipocyte-specificmarker expression in BAT (C andD) andWnuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI) (blue) (scale: 50 μm). The renormalized to β-actin (used as reference gene/protein). Data from the NCD group were set toANOVA and denoted as follows: &Pb.05, &&Pb.01, &&&Pb.001, HFD-G vs HFD; #Pb.05, ##Pb.01, ###Pdifference.

browning in adipocytes, such as PGC-1α, PRDM16 and CIDEA.Among them, PRDM16 is a determinant factor of brown-fat develop-ment and brings about the induction of BAT-specific genes byassociating with the transcriptional coactivators PGC-1α [43,44].PGC-1α controls respiration through induction of UCPs and regulationof nuclear respiratory factors [45]. In muscle-specific PGC-1αtransgenic mice, UCP1 and CIDEA mRNA expressions were up-regulated in WAT [46]. Meanwhile, adipose PGC-1α deficiency inmice results in a blunted expression of these thermogenic genes inWAT [47]. Furthermore, some beige adipocyte markers, includingTMEM26 and CITED1, also showed significant upregulation afterginger administration. These findings strongly suggested that gingercould activate the BAT thermogenic progress and induce adipocyte

. mRNA expression of brown and beige adipocyte-selective markers in BAT (A) andWATAT (E and F) (n=6); Immunofluorescence staining inWAT and BAT sections (G) (n=3);sults are expressed as the means±S.E.M. The ratios of specific mRNA or protein levels100%, and the rest of the values were normalized to this. Differences were assessed byb.001, HFD vs. NCD; ⁎Pb.05, ⁎⁎Pb.01, ⁎⁎⁎Pb.001, NCD-G vs NCD. Nomark= no significant

Page 8: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Fig. 4. (continued).

112 J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

Page 9: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

Fig. 4. (continued).

113J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

browning in WAT. Additionally, the thermogenesis of BAT is wellknown to be closely related to the number and function ofmitochondria. TFAM is a key regulator of mitochondrial replicationand biogenesis, and PGC-1α is the master regulator of mitochondrialbiogenesis by controlling the expression of TFAM by interacting withNRF1 [45]. Moreover, activation of PGC-1α plays an important role inmitochondrial biogenesis, and SIRT1-mediated PGC-1α deacetylationand activation are critical for mitochondrial function in BAT [48]. Inthis study, we found that ginger treatment couldmarkedly elevate theexpression of genes involved in mitochondrial biogenesis, such asPGC-1α, TFAM and NRF1, both in BAT and WAT. Moreover, theexpression of core proteins (SIRT1 and PGC-1α) were significantlyupregulated after ginger administration. Certainly, these resultsindicated that ginger supplementation could activate the thermogen-esis of BAT through upregulating mitochondrial biogenesis andfunction.

AMPK, a key regulator of energy metabolism, can affect theexpression of the key factors involved inmitochondrial biogenesis andenergy expenditure [49]. Furthermore, AMPK and SIRT1 act togetherwith PGC-1α to regulate energy homeostasis [8]. Activation of PGC-1αfurther upregulates the expression of PRDM16 and UCP1, thedominant regulator of energy expenditure [43]. Interestingly, anumber of phytochemicals (such as raspberry and resveratrol)promoteWAT browning and nonshivering thermogenesis by increas-ing expression and activation of AMPK and SIRT1, along with PGC-1α[40,50]. Consistent results in this study showed that ginger couldeffectively activate AMPK-promoted PGC-1α and SIRT1 proteinexpression and improve mitochondrial biogenesis, resulting inbrowning of WAT. These results indicated that ginger amelioratedHFD-induced obesity by promoting the browning of WAT andactivating the thermogenic program through the SIRT1/AMPK/PGC-1α signaling pathway in HFD-induced obese mice. Future studies onthe molecular mechanisms involved in energy metabolism regulationare required to elucidate the contribution of ginger to the preventionand treatment of obesity and related metabolic disorders. However,

due to the complex composition of ginger, its precise phytochemicalscontributing to its effects on energy metabolism remain unknown.Thus, more evidence is required to fully assess the bioactivesubstances in ginger that prevent obesity by regulating energymetabolism.

In conclusion, oral ginger supplementation significantly preventedand improved HFD-induced obesity, triggered the energy metabolismremodeling and enhanced browning-related gene and proteinexpression both in WAT and BAT. Briefly, ginger can regulate theglycolysis/gluconeogenesis-TCA cycle pathway and stimulate thebrowning program by the SIRT1/AMPK/PGC-1α pathway. Based onthese findings, we concluded that ginger is an edible plant that plays arole in the therapeutic treatment of obesity and related disorders.

Supplementary data to this article can be found online at https://doi.org/10.1016/j.jnutbio.2019.05.001.

Acknowledgments

This work was supported by the grant from National KeyTechnologies R&D Program [grant number 2017YFD0400700] andthe Beijing Municipal Science and Technology Project [grant numberD16110500540001].

Competing interests

The authors declare no competing interests.

Author contributions

J. W. designed and executed the experiments, analyzed the data,and wrote the manuscript; X-SH, P. W. and F. C. contributed to theexperimental design; D-TL contributed to performing experiments;All authors contributed to and have approved the final manuscript.

Page 10: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

114 J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

Abbreviations

BAT brown adipose tissueWAT white adipose tissueEpi-WAT epididymal white adipose tissuePer-WAT perirenal white adipose tissueIng-SAT inguinal subcutaneous adipose tissueNCD normal chow dietHFD high-fat dietNCD-G normal chow diet supplemented with gingerHFD-G high-fat diet supplemented with gingerTCA tricarboxylic acid cycleGC–MS gas chromatography–mass spectrometryACTB beta-actinSIRT1 sirtuin-1AMPK AMP-activated protein kinasePGC-1α peroxisome proliferator-activated receptorγ coactivator 1αUCP1 uncoupling protein-1CIDEA Cell death-inducing DNA fragmentation factor alpha-like

effector ANRF-1 nuclear respiratory factor-1TFAM transcription factor APPARs peroxisome proliferator-activated receptorsGLU glucoseTC total cholesterolTG triglycerideLDL low-density lipoproteinHDL high-density lipoproteinALT alanine aminotransferaseAST aspartate aminotransferaseRER respiratory exchange ratio

References

[1] Smith KB, Smith MS. Obesity statistics. Prim Care 2016;43:121. https://doi.org/10.1016/j.pop.2015.10.001.

[2] Chen LH, Chen YH, Cheng KC, Chien TY, Chan CH, Tsao SP, et al. Antiobesity effectof lactobacillus reuteri 263 associated with energy metabolism remodeling ofwhite adipose tissue in high-energy-diet-fed rats. J Nutr Biochem 2017;54:87–94.https://doi.org/10.1016/j.jnutbio.2017.11.004.

[3] Cypess AM, Lehman S, Williams G, Tal I, Rodman D, Goldfine AB, et al.Identification and importance of brown adipose tissue in adult humans. N EnglJ Med 2009;360:1509–17. https://doi.org/10.1056/NEJMoa0810780.

[4] Peirce V, Carobbio S, Vidalpuig A. The different shades of fat. Nature 2014;510:76–83. https://doi.org/10.1038/nature13477.

[5] Townsend KL, Tseng YH. Brown fat fuel utilization and thermogenesis. TrendsEndocrinol Metab 2014;25:168–77. https://doi.org/10.1016/j.tem.2013.12.004.

[6] Wu J, Cohen P, Spiegelman BM. Adaptive thermogenesis in adipocytes: is beigethe new brown? Genes Dev 2013;27:234–50. https://doi.org/10.1101/gad.211649.112.

[7] Suwa M, Nakano H, Kumagai S. Effects of chronic AICAR treatment on fibercomposition, enzyme activity, UCP3, and PGC-1 in rat muscles. J Appl Physiol2003;95:960–8. https://doi.org/10.1152/japplphysiol.00349.2003.

[8] Cantó C, Auwerx J. PGC-1alpha, SIRT1 and AMPK, an energy sensing network thatcontrols energy expenditure. Curr Opin Lipidol 2009;20:98–105. https://doi.org/10.1097/MOL.0b013e328328d0a4.

[9] Baboota RK, Murtaza N, Jagtap S, Singh DP, Karmase A, Kaur J, et al. Capsaicin-induced transcriptional changes in hypothalamus and alterations in gut microbialcount in high fat diet fed mice. J Nutr Biochem 2014;25:893–902. https://doi.org/10.1016/j.jnutbio.2014.04.004.

[10] Lone J, Choi JH, Kim SW, Yun JW. Curcumin induces brown fat-like phenotype in3T3-L1 and primary white adipocytes. J Nutr Biochem 2015;27:193–202. https://doi.org/10.10516/j.jnutbio.2015.09.006.

[11] Kowalska K, Olejnik A, Rychlik J, Grajek W. Cranberries (Oxycoccus quad-ripetalus) inhibit lipid metabolism and modulate leptin and adiponectinsecretion in 3T3-L1 adipocytes. Food Chem 2015;185:383–8. https://doi.org/10.1016/j.foodchem.2015.03.152.

[12] Cavalera M, Axling U, Berger K, Holm C. Rose hip supplementation increasesenergy expenditure and induces browning of white adipose tissue. Nutr Metab(Lond) 2016;13:e91. https://doi.org/10.1186/s12986-016-0151-5.

[13] Oh S, Lee MS, Jung S, Kim S, Park H, Park S, et al. Ginger extract increases musclemitochondrial biogenesis and serum HDL-cholesterol level in high-fat diet-fedrats. J Funct Foods 2017;29:193–200. https://doi.org/10.1016/j.jff.2016.12.023.

[14] Li Y, Hong Y, Han Y, Wang Y, Xia L. Chemical characterization and antioxidantactivities comparison in fresh, dried, stir-frying and carbonized ginger. JChromatogr B Analyt Technol Biomed Life Sci 2016;1011:223–32. https://doi.org/10.1016/j.jchromb.2016.01.009.

[15] Wang J, Ke W, Bao R, Hu X, Chen F. Beneficial effects of ginger Zingiber officinaleroscoe on obesity and metabolic syndrome: a review. Ann N Y Acad Sci 2017;1398:83–98. https://doi.org/10.1111/nyas.13375.

[16] 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 theperoxisome proliferator-activated receptor δ pathway. J Nutr Biochem 2015;26:1058–67. https://doi.org/10.1016/j.jnutbio.2015.04.014.

[17] Wang J, Gao H, Ke D, Zuo G, Yang Y, Yamahara J, et al. Improvement of liquidfructose-induced adipose tissue insulin resistance by ginger treatment in rats isassociated with suppression of adipose macrophage-related proinflammatorycytokines. Evid Based Complement Alternat Med 2013;2013:e590376. https://doi.org/10.1155/2013/590376.

[18] Brahmanaidu P, Uddandrao VV, Ravindarnaik R, Suresh P, Meriga B, BegumMS,et al. Ameliorative potential of gingerol: promisingmodulation of inflammatoryfactors and lipid marker enzymes expressions in HFD induced obesity in rats.Mol Cell Endocrinol 2015;419:139–47. https://doi.org/10.1016/j.mce.2015.10.007.

[19] Saravanan G, Ponmurugan P, Deepa MA, Senthilkumar B. Anti-obesity action ofgingerol: effect on lipid profile, insulin, leptin, amylase and lipase in male obeserats induced by a high-fat diet. J Sci Food Agric 2014;94:2972–7. https://doi.org/10.1002/jsfa.6642.

[20] Tzeng TF, Liu IM. 6-gingerol prevents adipogenesis and the accumulation ofcytoplasmic lipid droplets in 3T3-L1 cells. Phytomedicine 2013;20:481–7. https://doi.org/10.1016/j.phymed.2012.12.006.

[21] Suk S, Seo SG, Yu JG, Yang H, Jeong E, Jang YJ, et al. A bioactive constituent ofginger, 6-shogaol, prevents adipogenesis and stimulates lipolysis in 3T3-L1adipocytes. J Food Biochem 2016;40:84–90. https://doi.org/10.1111/jfbc.12191.

[22] Matsuda A, Wang ZZ, Takahashi S, Tokuda T, Miura N, Hasegawa J. Upregulation ofmRNA of retinoid binding protein and fatty acid binding protein by cholesterolenriched-diet and effect of ginger on lipid metabolism. Life Sci 2009;84:903–7.https://doi.org/10.1016/j.lfs.2009.04.004.

[23] Zhang X, Zhang QX, Wang X, Zhang L, QuW, Bao B, et al. Dietary luteolin activatesbrowning and thermogenesis in mice through an AMPK/PGC1α pathway-mediated mechanism. Int J Obes (Lond) 2016;40:1841–9. https://doi.org/10.1038/ijo.2016.108.

[24] Gao H, Guan T, Li C, Zuo G, Yamahara J, Wang J, et al. Treatment with gingerameliorates fructose-induced fatty liver and hypertriglyceridemia in rats:modulation of the hepatic carbohydrate response element-binding protein-mediated pathway. Evid Based Complement Alternat Med 2012;2012:570948.https://doi.org/10.1155/2012/570948.

[25] Li Y, Tran VH, Kota BP, Nammi S, Duke CC, Roufogalis BD. Preventative effect ofZingiber officinale on insulin resistance in a high-fat high-carbohydrate diet-fedrat model and its mechanism of action. Basic Clin Pharmacol Toxicol 2014;115:209–15.

[26] Patel DP, Krausz KW, Xie C, Beyoğlu D, Gonzalez FJ, Idle JR. Metabolic profiling bygas chromatography-mass spectrometry of energy metabolism in high-fat diet-fed obese mice. Plos One 2017;12:e0177953. https://doi.org/10.1371/journal.pone.0177953.

[27] Mediani A, Abas F, Maulidiani M, Abu Bakar Sajak A, Khatib A, Tan CP, et al.Metabolomic analysis and biochemical changes in the urine and serum ofstreptozotocin-induced normal- and obese-diabetic rats. J Physiol Biochem 2018;74:403–16. https://doi.org/10.1007/s13105-018-0631-3.

[28] So-Hyun K, Seung-Ok Y, Hee-Su K, Yujin K, Taesun P, Hyung-Kyoon C. 1H-nuclearmagnetic resonance spectroscopy-based metabolic assessment in a rat model ofobesity induced by a high-fat diet. Anal Bioanal Chem 2009;395:1117–24. https://doi.org/10.1007/s00216-009-3054-8.

[29] Sun JH, Liu X, Cong LX, Li H, Zhang CY, Chen JG, et al. Metabolomics study of thetherapeutic mechanism of Schisandra Chinensis lignans in diet-induced hyperlip-idemia mice. Lipids Health Dis 2017;16:e145. https://doi.org/10.1186/s12944-017-0533-3.

[30] Heinrichsen ET, Hui Z, Robinson JE, John N, Soda D, Rolf B, et al. Metabolic andtranscriptional response to a high-fat diet in Drosophila melanogaster. Mol Metab2014;3:42–54. https://doi.org/10.1016/j.molmet.2013.10.003.

[31] Carrière A, Jeanson Y, Bergermüller S, André M, Chenouard V, Arnaud E, et al.Browning of white adipose cells by intermediate metabolites: an adaptivemechanism to alleviate redox pressure. Diabetes 2014;63:3253–65. https://doi.org/10.2337/db13-1885.

[32] Jambocus NGS, Saari N, Ismail A, Khatib A, Mahomoodally MF, Hamid AA. Aninvestigation into the antiobesity effects of Morinda citrifolia L. leaf extract inhigh fat diet induced obese rats using a H-1 NMR metabolomics approach. JDiabetes Res 2016;27:e2391592. https://doi.org/10.1155/2016/2391592.

[33] Shearer J, Duggan G, Weljie A, Hittel DS, Wasserman DH, Vogel HJ. Metabolomicprofiling of dietary-induced insulin resistance in the high fat-fed C57BL/6J

Page 11: Ginger prevents obesity through regulation of energy ......[2]. Adipose tissue is a major metabolic tissue and plays a key role in energy homeostasis. Currently, three types of adipose

115J. Wang et al. / Journal of Nutritional Biochemistry 70 (2019) 105–115

mouse. Diabetes Obes Metab 2008;10:950–8. https://doi.org/10.1111/j.1463-1326.2007.00837.x.

[34] Jiang CY, Yang KM, Yang L, Miao ZX, Wang YH, Zhu HB. A 1H NMR-basedmetabonomic investigation of time-related metabolic trajectories of the plasma,urine and liver extracts of hyperlipidemic hamsters. Plos One 2013;8:e66786.https://doi.org/10.1371/journal.pone.0066786.

[35] Ricquier D. Fundamental mechanisms of thermogenesis. C R Biol 2006;329:578–86. https://doi.org/10.1016/j.crvi.2005.10.010.

[36] You Y, Yuan X, Liu X, Liang C, Meng M, Huang Y, et al. Cyanidin-3-glucosideincreases whole body energy metabolism by upregulating brown adipose tissuemitochondrial function. Mol Nutr Food Res 2017;61:1700261. https://doi.org/10.1002/mnfr.201700261.

[37] Ricquier D, Bouillaud F, Toumelin P, Mory G, Bazin R, Arch J, et al. Expression ofuncoupling protein mRNA in thermogenic or weakly thermogenic brown adiposetissue. Evidence for a rapid beta-adrenoreceptor-mediated and transcriptionallyregulated step during activation of thermogenesis. J Biol Chem 1986;261:13905–10. https://doi.org/10.1109/I2MTC.2014.6861005.

[38] Tao H, Yuan X, Gang W, Luo H, Lee HJ, Jin W. Myricetin-induced brown adiposetissue activation prevents obesity and insulin resistance in db/db mice. Eur J Nutr2018;57:391–403. https://doi.org/10.1007/s00394-017-1433-z.

[39] Seo YJ, Kim KJ, Jia C, Koh EJ, Lee BY. Spirulina maxima extract reduces obesitythrough suppression of adipogenesis and activation of browning in 3T3-L1 cellsand high-fat diet-induced obese mice. Nutrients 2018;10:e712. https://doi.org/10.3390/nu10060712.

[40] Zou T, Chen D, Yang Q, Wang B, Zhu MJ, Nathanielsz PW, et al. Resveratrolsupplementation of high-fat diet-fed pregnant mice promotes brown and beigeadipocyte development and prevents obesity in male offspring. J Physiol 2017;595:1547–62. https://doi.org/10.1113/JP273478.

[41] Liu Z, Qiao Q, Sun Y, Chen Y, Ren B, Liu X. Sesamol ameliorates diet induced obesityin C57BL/6J mice and suppresses adipogenesis in 3T3-L1 cells via regulatingmitochondria-lipid metabolism. Mol Nutr Food Res 2017;61:e1600717. https://doi.org/10.1002/mnfr.201600717.

[42] Zuo J, Zhao D, Yu N, Fang X, Mu Q, Ma Y, et al. Cinnamaldehyde ameliorates diet-induced obesity in mice by inducing browning of white adipose tissue. CellPhysiol Biochem 2017;42:1514–25. https://doi.org/10.1159/000479268.

[43] Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls abrown fat/skeletal muscle switch. Nature 2008;454:961–7. https://doi.org/10.1038/nature07182.

[44] Shingo K, Patrick S, Takuya T, Hediye EB, Cooper MP, Ruas JL, et al. Regulation ofthe brown and white fat gene programs through a PRDM16/CtBP transcrip-tional complex. Genes Dev 2008;22:1397–409. https://doi.org/10.1101/gad.1666108.

[45] Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, et al.Mechanisms controlling mitochondrial biogenesis and respiration through thethermogenic coactivator PGC-1. Cell 1999;98:115–24. https://doi.org/10.1016/S0092-8674(00)80611-X.

[46] Pontus BM, Jun W, Jedrychowski MP, Anisha K, Li Y, Lo JC, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat andthermogenesis . Nature 2013;481:463–8. https://doi .org/10.1038/nature10777.

[47] Sandra K, Mepani RJ, Dina L, Li Y, Jurczak MJ, Jornayvaz FR, et al. Development ofinsulin resistance inmice lacking PGC-1α in adipose tissues. Proc Natl Acad Sci U SA 2012;109:9635–40. https://doi.org/10.1073/pnas.1207287109.

[48] Rodgers JT, Carlos L, Wilhelm H, Gygi SP, Spiegelman BM, Pere P. Nutrient controlof glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 2005;434:113–8. https://doi.org/10.1038/nature03354.

[49] Lu Y, Wu Q, Liu LZ, Yu XJ, Liu JJ, Li MX, et al. Pyridostigmine protects againstcardiomyopathy associated with adipose tissue browning and improvement ofvagal activity in high-fat diet rats. Biochim Biophys Acta Mol Basis Dis 2018:1037–50. https://doi.org/10.1016/j.bbadis.2018.01.006.

[50] Wang S, Liang X, Yang Q, Fu X, Zhu M, Rodgers BD, et al. Resveratrol enhancesbrown adipocyte formation and function by activating AMP-activated proteinkinase (AMPK) α1 in mice fed high-fat diet. Mol Nutr Food Res 2016;61:e1600746. https://doi.org/10.1002/mnfr.201600746.


Recommended