You are on page 1of 10

FOOD & FUNCTION

Pears www.mnf-journal.com

Pear Extract and Malaxinic Acid Reverse Obesity, Adipose


Tissue Inflammation, and Hepatosteatosis in Mice
Xuan T. Truong, Thuy T. P. Nguyen, Man-Jong Kang, Chang Hwa Jung, Sueun Lee,
Changjong Moon, Jae-Hak Moon, and Tae-Il Jeon*

Scope: Obesity and diabetes are major public health problems and are 1. Introduction
emerging as pandemics. Considerable evidence suggests that pear fruit The increasing prevalence of obesity is a
consumption is associated with a lower risk of obesity-related complications. global threat to public health. It is esti-
Thus, the present study is conducted to investigate the therapeutic potential mated that there were more than 2 bil-
lion obese or overweight people world-
of pear extract (PE) for reversing obesity and associated metabolic
wide in 2013, which represents 30% of
complications in high-fat diet-induced obese mice. the world’s population.[1] The economic
Methods and results: Obesity is induced in male C57BL/6 mice fed a high-fat cost related to obesity and its complica-
diet for 11 weeks. After the first 6 weeks on the diet, obese mice are tions is very high.[2]
administered vehicle or PE for 5 weeks. PE treatment decreases body weight Massive expansion of white adipose
gain, expands white adipose tissue (WAT), and causes hepatic steatosis in tissue (WAT), a hallmark of obesity, is
characterized by adipocyte hypertrophy
obese mice, as well as inhibits adipogenesis and lipogenesis. Impaired and hyperplasia. This phenomenon leads
glucose tolerance and insulin resistance are improved by PE. In addition, PE to dysfunction of adipose tissue accom-
reduces macrophage infiltration and expression of pro-inflammatory genes panied by increased infiltration of im-
and deactivates mitogen-activated protein kinases in WAT. Finally, malaxinic mune cells, particularly macrophages,
acid is identified as an active component responsible for the anti-obesity and the activation of inflammation and
is largely associated with obesity-related
effects of PE in mice.
complications such as insulin resistance
Conclusion: The results demonstrate that PE supplementation ameliorates (IR), type 2 diabetes (T2D), and non-
diet-induced obesity and associated metabolic complications and suggest the alcoholic fatty liver disease (NAFLD).[3]
health-beneficial effects of both pear fruits and malaxinic acid in WAT inflammation in obesity can pro-
counteracting these diseases. mote IR, which enhances the lipoly-
sis of stored fat. Increased delivery of
fatty acids to the liver results in exces-
sive accumulation of intracellular lipids,
particularly triglyceride, and the subsequent development of
X. T. Truong, T. T. P. Nguyen, Prof. M.-J. Kang, Prof. T.-I. Jeon NAFLD.[4] Indeed, several clinical studies have shown that WAT
Department of Animal Science
Chonnam National University inflammation in obese patients with NAFLD was greater than
Gwangju 61186, Republic of Korea that in obese patients with normal hepatic triglyceride levels.[5]
E-mail: tjeon@jnu.ac.kr Therefore, modulating processes that control the expansion or
Dr. C. H. Jung differentiation of WAT may be an effective strategy for prevent-
Research Group of Natural Materials and Metabolism ing or treating metabolic diseases, including obesity.
Korea Food Research Institute
Intensive lifestyle modification including diet and exercise is
Wanju-gun, Jeollabuk-do 55365, Republic of Korea
clinically beneficial for many obese patients. However, changing
S. Lee, Prof. C. Moon
Department of Veterinary Anatomy and Animal Behavior behavioral factors is difficult to achieve and maintain and phar-
College of Veterinary Medicine and BK21 Plus Project Team macological options are often required. Although several anti-
Chonnam National University obesity agents are currently available, long-term consumption of
Gwangju 61186, Republic of Korea these agents may have severe side effects.[6] Increasing evidence
Prof. J.-H. Moon indicates that fruit consumption is associated with a lower risk of
Department of Food Science and Technology and Functional Food
Research Center obesity-related complications, and many national guidelines rec-
Chonnam National University ommend increasing fruit and vegetable consumption to manage
BK21 Plus Program, Gwangju 61186, Republic of Korea overall health and obesity.[7]
Pears (Pyrus spp.) are commonly consumed worldwide in ei-
The ORCID identification number(s) for the author(s) of this article ther fresh or processed forms such as juices and jams in daily
can be found under https://doi.org/10.1002/mnfr.201801347 life and are also traditionally used as herbal medicines to treat
DOI: 10.1002/mnfr.201801347 respiratory diseases in Asia.[8] Our and other research groups

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (1 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

previously showed that pear fruits, particularly those that are im- treating confluent cells for 48 h in medium containing 10% FBS,
mature, are rich sources of bioactive phenolic compounds with 0.5 mm IBMX, 1 µm DEX, and 10 µg mL−1 insulin. The medium
antioxidant and anti-inflammatory properties.[8b,9] Recently, di- was replaced with that supplemented with only 5 µg mL−1 insulin
etary fruit polyphenols have gained recognition as promising every other day. The cells were treated with or without PE or MA
agents for conferring protection against metabolic diseases;[10] for 8 days during adipogenesis.
two clinical trials have indeed suggested that increased daily pear
consumption is inversely correlated with weight gain.[11] Thus, in
the current study, we examined whether pear can improve high- 2.4. Oil Red O Staining
fat diet (HFD)-induced obesity. We found that pear treatment
effectively improved HFD-induced body weight gain, IR, and Cells were fixed with 10% formalin solution for 10 min and
hepatic steatosis in mice. Pear predominantly targeted adipose stained with freshly prepared Oil Red O solution for 30 min at
tissue, thereby reducing adipocyte hypertrophy and WAT inflam- RT. Images were acquired using a Leica DM IL LED microscope
mation. Moreover, we showed that malaxinic acid (MA) is a po- (Wetzlar, Germany). Intracellular lipid contents were measured
tent anti-obesity component in pear. by extracting Oil Red O with isopropanol, and the absorbance was
measured at 500 nm with a microplate reader (Biotek, Winooski,
VT).
2. Experimental Section
2.1. Materials and Sample Preparation 2.5. RNA Analysis

DMEM, fetal calf serum (FCS), fetal bovine serum (FBS), and an- Total RNA from tissues and cells was isolated using TRI-
tibiotics were purchased from Hyclone (Logan, UT). 3-Isobutyl-1- zol(Invitrogen, Carlsbad, CA) and cDNA was synthesized using a
methylxanthine (IBMX), dexamethasone (DEX), insulin, and Oil ReverTra Ace qPCR RT Kit (Toyobo, Osaka, Japan). Quantitative
red O powder were obtained from Sigma-Aldrich (St. Louis, MO). PCR was performed with SYBR Green Master Mix (Enzynomics,
The whole immature pear fruits (2.5 kg fresh wt.) were pulverized Daejeon, South Korea) using a Mx3000P real-time PCR System
using a square cutter, and extracted with 25 L of hot water (2 h, (Agilent Technologies, Santa Clara, CA). Primer sequences are
100 °C), followed by filtration to obtain an extract. The extract shown in Table S1, Supporting Information. mRNA levels were
was concentrated in a vacuum at 60 °C, lyophilized for 3 days, normalized for expression to mouse ribosomal protein, Large, P0
and stored at −20 °C until use. The detailed methods for isola- (RPLP0) as control and calculated by the comparative threshold
tion, purification, and identification of MA from pear extract (PE) cycle method.
were reported in our previous studies.[9a,12]

2.6. Immunoblotting
2.2. Analysis of Polyphenol Composition in PE
Cells and tissues were lysed in RIPA buffer containing protease
Total phenolic and flavonoid contents in PE were measured using and phosphatase inhibitors. The lysates were sonicated and cen-
Folin–Ciocalteu’s method and the aluminum chloride colorimet- trifuged at 20 000 × g for 20 min at 4 °C. The supernatants were
ric assay, respectively[8a] (Figure S1A, Supporting Information). separated by SDS-PAGE and then transferred to nitrocellulose
For polyphenol composition analysis, PE was dissolved in 80% membranes. The membranes were incubated with primary anti-
MeOH and subjected to HPLC using an ODS column (UG120, bodies (Table S2, Supporting Information) overnight at 4 °C, fol-
4.6 mm I.D. × 250 mm, 5 µm, Shiseido, Tokyo, Japan). The mo- lowed by incubation with secondary antibodies (Thermo Fisher
bile phase was composed of H2 O:acetic acid (98:2, v/v, eluent A) Scientific, Waltham, MA). Blots were developed using an EZ-
and 50% MeOH (eluent B). The gradient program began at 100% Western Lumi Pico or Femto western blot detection kit (DAEIL-
A and was increased to 100% B over a period of 40 min. The col- LAB Service, Seoul, South Korea).
umn temperature was maintained at 40 °C, and the flow rate was
1 mL min−1 . Phenolic compounds were monitored at 280 and
254 nm, respectively, using a photodiode array detector (SPD- 2.7. Animals
M20D; Shimadzu, Kyoto, Japan). The contents were quantified
against the chromatographic peak areas of the external standards. All animal studies were performed in accordance with accepted
The calibration curves were plotted over a concentration range of standards of animal welfare and with permission from the
0.01–10 µg (Figure S1, Supporting Information). Chonnam National University Institutional Animal Care and
Use Committee (YB-2016-01). Five-week-old male C57BL/6 mice
were obtained from OrientBio (Seongnam, South Korea) and
2.3. Cell Culture maintained on a chow diet for 1 week with a 12 h light/dark cycle
for acclimatization. At 6 weeks of age, the mice were randomly
3T3-L1 cells were purchased from the American Type Culture assigned (n = 5 per group) to a group fed regular chow diet (CD,
Collection (Manassas, VA). Cells were maintained in DMEM sup- LabDiet 5L79, OrientBio) or to a group fed HFD (Research Diets,
plemented with 10% FCS and 1% antibiotics in an atmosphere #D12492, New Brunswick, NJ). The CD provided 3.43 kcal g−1
of 5% CO2 at 37 °C. Adipocyte differentiation was induced by of energy (21% calories from protein, 14% calories from fat, and

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (2 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

65% calories from carbohydrate), whereas the HFD provided 5.21 and then antigen retrieval was performed by heating the sections
kcal g−1 of energy (20% calories from protein, 60% calories from with 10 mm citrate buffer, pH 6. Sections were blocked in 5%
fat, and 20% calories from carbohydrate). The ingredients includ- normal goat serum and 0.1% Tween 20, reacted with anti-IBA1
ing sucrose in CD were not perfectly matched with HFD. After 6 (1:400) overnight at 4 °C, and then incubated with biotinylated
weeks on the diet, HFD-induced obese mice were orally admin- goat anti-rabbit IgG (Vector Laboratories, Burlingame, CA) for 1 h
istered with vehicle (saline) or PE at 200 mg per kg body weight at RT. The immunoreaction with avidin–biotin peroxidase com-
per day for 5 weeks. In MA analysis, mice were fed a HFD for 9 plex (Vector Laboratories) was conducted for 1 h at RT. The per-
weeks, followed by oral administration of MA (40 mg per kg body oxidase reaction was developed using a DAB Substrate Kit (Vec-
weight per day) for 7 weeks. Body weight was measured weekly tor Laboratories). Images were acquired with a BX-40 microscope
and food intake was recorded every other day. After the feeding (Olympus, Tokyo, Japan) fitted with an eXcope × 3 digital camera
periods, the mice were sacrificed at 8 a.m. (at the end of the dark (DIXI Optics, Daejeon, South Korea). Average adipocyte size, fre-
cycle) by CO2 asphyxiation. quency distribution of adipocyte sizes, and number of crown-like
structures (CLSs) were counted using Image J software.

2.8. Dosage Information


2.12. Statistical Analysis
Mice were orally gavaged with PE (200 mg kg body weight−1 )
or MA (40 mg kg body weight−1 ) once daily for 5 or 9 weeks, Data were presented as the mean ± SEM. Differences between
respectively. The dosages were chosen according to the previ- the means of individual groups were assessed by one-way analy-
ous studies.[[13] The dose of PE or MA used in the present study sis of variance followed with Tukey’s multiple comparisons test;
was roughly equivalent to 16.2 or 3.2 mg kg body weight−1 in differences were considered significant at p < 0.05. The statisti-
humans,[14] which equates to a 972-mg dose of PE or a 192-mg cal software package, Prism 6.0 (GraphPad Software, La Jolla, CA,
dose of MA for a person weighing 60 kg. Based on an average of USA), was used for these analyses.
0.5 g PE derived from each immature pear fruit (10 g), an indi-
vidual should consume approximately two immature pear fruits
per day. 3. Results
3.1. PE Reduces Body Weight Gain and Improves Glucose
2.9. Biochemical Analysis of Plasma Homeostasis in HFD-Induced Obese Mice

Blood was collected by cardiac puncture and the plasma was sepa- Six weeks of HFD feeding resulted in significantly increased body
rated by centrifugation. Plasma glucose, triglyceride, total choles- weight and impaired glucose tolerance compared with that in CD
terol, glutamic oxaloacetic transaminase, and glutamic pyruvic mice (data not shown). To investigate the therapeutic effects of PE
transaminase were determined using Asan enzymatic kits (Asan, on diet-induced obesity, we orally administered vehicle (saline)
Seoul, South Korea). Plasma insulin concentrations were mea- or PE to HFD-induced obese mice for 5 weeks. Although food in-
sured with the Ultrasensitive mouse Insulin ELISA Kit (ALPCO, take was similar as in the vehicle control group, PE supplemen-
Salem, NH). The homeostasis model assessment for insulin re- tation significantly decreased body weight (Figure 1A,B). Next,
sistance (HOMA-IR), an indicator of IR, was calculated with a to determine the effect of PE on glucose homeostasis and in-
HOMA2 calculator released by the Diabetes Trials Unit, Univer- sulin sensitivity, GTT and ITT were performed after 4 weeks of
sity of Oxford.[15] PE treatment. As shown in Figure 1C,D, PE-treated HFD-fed
mice exhibited lower glucose levels at all time points up to 120
min after oral glucose challenge or intraperitoneal insulin injec-
2.10. Glucose and Insulin Tolerance Test tion and reduced areas under the curves compared with vehicle
control HFD-fed mice. Moreover, PE supplementation lowered
For the glucose tolerance test (GTT), mice were fasted for 16 h the plasma levels of glucose and insulin, thereby decreasing the
prior to oral gavage of glucose (1 g kg body weight−1 ). In the in- HOMA-IR index (Figure 1E). Additionally, PE supplementation
sulin tolerance test (ITT), mice were intraperitoneally injected significantly decreased the plasma levels of triglyceride and total
with insulin (0.75 U kg body weight−1 ) after fasting for 4 h. cholesterol, whereas no differences were observed in enzymes
The glucose concentrations were measured at 0, 15, 30, 60, 90, associated with hepatic injury (Figure S2, Supporting Informa-
and 120 min in the tail blood using a glucometer (Roche, Basel, tion). These results indicate that PE treatment effectively reduced
Switzerland). obesity-associated hyperglycemia, hyperinsulinemia, and hyper-
lipidemia in mice, and these effects did not result from reduced
food consumption or toxicity.
2.11. Histology and Immunohistochemistry

Tissues were fixed with 4% paraformaldehyde, embedded in 3.2. PE Decreases WAT Expansion by Inhibiting Adipogenesis
paraffin wax, and sectioned sagittally (4 µm thick). Multiple sec-
tions were stained with hematoxylin and eosin to detect mor- In parallel with weight loss caused by PE, WAT weights (Fig-
phological changes. The sections were dewaxed and hydrated, ure 2A), mean adipocyte size (Figure 2B), and large adipocyte

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (3 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

Figure 1. PE promotes weight loss and improves glucose homeostasis and insulin sensitivity in HFD-induced obese mice. Mice were fed a HFD for 6
weeks. Subsequently the same mice were administered with vehicle or PE (200 mg kg−1 day−1 ) on the diet for an additional 5 weeks. A) Body weight
gain. B) Food intake. C) GTT and D) ITT were performed at 10 weeks. E) Fasting plasma glucose and insulin levels. HOMA-IR was determined using
both levels. Data are mean ± SEM; n = 5. Data are representative of two independent experiments with five mice per group. *p < 0.05 versus CD; # p <
0.05 versus HFD (vehicle control).

numbers (Figure 2C) were remarkably reduced. Activation of en- in vitro (Figure 3C,D). Interestingly, PE also restored HFD dys-
ergy expenditure in brown adipose tissue (BAT) and WAT brown- regulation of the adipokine leptin and adiponectin expression in
ing has recently emerged as a promising strategy for treating WAT (Figure 2D). These results suggest that reduced adiposity
metabolic diseases.[16] However, the expression of thermogenic by PE is associated with decreased adipocyte hypertrophy and in-
gene uncoupling protein 1 (UCP1) was not changed in both hibition of adipogenesis.
BAT and inguinal WAT from PE-treated mice (Figure S3A, Sup-
porting Information), suggesting that the reduced body weight
gain resulted from inhibition of fat accumulation in WAT. To 3.3. PE Prevents Hepatic Steatosis in HFD-Induced Obese Mice
evaluate this hypothesis, murine preadipocyte 3T3-L1 cells were
differentiated in the presence or absence of PE. As shown in Adipocyte dysfunction in obesity influences ectopic fat accumu-
Figure 3A,B, PE significantly suppressed fat accumulation as as- lation and IR in the liver, leading to NAFLD.[17] We therefore ex-
sessed by Oil Red O staining in a dose-dependent manner. Fur- amined the effects of PE on hepatic steatosis. Although there
thermore, the expression of adipogenic transcription factors such was no significant difference in liver weights between groups,
as CCAAT/enhancer binding protein α (C/EBPα) and peroxi- histological examination revealed an increase in lipid droplets in
some proliferator-activated receptor γ (PPARγ ) and lipogenic the liver of HFD-induced obese mice, whereas PE supplementa-
genes was markedly inhibited by PE both in vivo (Figure 2D) and tion significantly attenuated fat accumulation (Figure 2E). This

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (4 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

Figure 2. PE reduces adiposity and hepatic steatosis. A) Tissue weights. B) H&E staining in WAT of mice (magnification: ×200, scale bar: 50 µm). C)
Average adipocyte area and frequency distribution of adipocyte sizes were counted using ImageJ. D) mRNA levels in WAT were analyzed by RT-qPCR. E)
H&E staining in liver of mice. F) Liver triglyceride levels. Data are mean ± SEM; n = 5. *p < 0.05 versus CD; # p < 0.05 versus HFD. eWAT, epididymal;
iWAT, inguinal; iBAT, interscapular.

observation was clearly confirmed by measuring hepatic triglyc- we next examined the effects of PE on HFD-induced WAT in-
eride levels (Figure 2F). We further analyzed the mRNA expres- flammation. Immunostaining of the macrophage-specific pro-
sion levels of genes involved in hepatic lipid metabolism. Com- tein IBA1 revealed abundant CLSs formed from clusters of
pared with the vehicle control mice, PE significantly suppressed macrophages surrounding necrotic adipocytes in the WAT of
lipogenic genes including sterol regulatory element binding HFD-fed mice, whereas PE markedly decreased the recruitment
protein-1c, stearoyl-CoA desaturase 1, fatty acid synthase, and of macrophages and the number of CLSs (Figure 4A). Consis-
acetyl-CoA carboxylase 1 (Figure S3B, Supporting Information). tent with these histological observations, after PE treatment, the
However, HFD suppression of carnitine palmitoyltransferase 1α mRNA expression levels of the macrophage-specific markers
for fatty acid oxidation was not recovered by PE (Figure S3B, Sup- F4/80 and Cd68 were significantly reduced (Figure 4B). Simi-
porting Information), suggesting that PE ameliorates obesity- larly, PE-treated mice showed reduced expression levels of sev-
associated hepatic steatosis by inhibiting lipogenesis. Together, eral markers of pro-inflammatory M1 macrophages including tu-
these results demonstrate that PE supplementation greatly re- mor necrosis factor α (Tnfα), monocyte chemoattractant protein
duced adiposity, maintained glucose homeostasis, and improved 1 (Mcp1), and integrin, alpha X (Itgax/Cd11c), whereas there were
hepatic steatosis in HFD-induced obese mice. no significant changes in the expression of the anti-inflammatory
M2 macrophage markers between groups (Figure 4C). While
mitogen-activated protein kinases (MAPKs) are essential for reg-
3.4. PE Suppresses HFD-Induced Macrophage Infiltration and ulating the proliferation and differentiation of adipocytes, ex-
Inflammation in WAT cessive activation of MAPKs is associated with metabolic dys-
functions in obesity and diabetes.[19] Thus, we examined the
As adipocyte hypertrophy is closely associated with WAT in- phosphorylation (activation) level of three major MAPKs, c-Jun
flammation, characterized by increased infiltration and polariza- N-terminal kinase (JNK), extracellular signal-regulated kinase
tion of macrophages to the pro-inflammatory M1 phenotype,[18] (ERK), and p38 in WAT. HFD feeding significantly increased the

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (5 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

Figure 3. PE inhibits adipogenesis in 3T3-L1 cells. 3T3-L1 cells were differentiated for 8 days in the presence or absence of PE. A) Oil Red O staining of
differentiated adipocytes and B) quantification via absorbance measurements. C,D) mRNA and protein expression levels were analyzed by RT-qPCR and
immunoblotting, respectively. Data are mean ± SEM; n = 3 for three separate experiments. *p < 0.05 versus preadipocyte; # p < 0.05 versus adipocyte
(vehicle control); ## p < 0.01 versus adipocyte. MDI, methylisobutylxanthine, dexamethasone, insulin; ORO, Oil Red O.

phosphorylation of all three MAPKs in the WAT, which was sig- hicle control group (Figure 5B). GTT and ITT revealed that MA-
nificantly reversed by PE treatment (Figure 4D). Macrophage- treated mice were significantly more glucose tolerant (Figure 5C)
specific JNK deficiency prevented the accumulation of adipose and sensitive to insulin (Figure 5D), with significantly lower fast-
tissue macrophages (ATMs) expressing surface markers for M1 ing plasma glucose and insulin levels also observed in these mice
polarization in the WAT of HFD-fed mice without affecting M2 (Figure S4, Supporting Information), suggesting that MA amelio-
polarization.[20] Therefore, our results indicate that PE decreased rated glucose homeostasis and IR in HFD-induced obese mice.
WAT inflammation in HFD-fed mice by reducing macrophage Consistent with the effects of PE on adiposity (Figure 2) and
infiltration and the number of ATMs with M1 phenotype in WAT. adipogenesis (Figure 3), MA treatment significantly reduced the
This suggests that deactivation of the JNK signaling pathway WAT mass (data not shown) and adipocyte differentiation accom-
contributes to ATM polarization by PE, although the regulatory panied by downregulation of PPARγ and C/EBPα both in vitro
mechanism remains unclear. (Figure S5, Supporting Information) and in vivo (Figure 5E). We
also observed a dramatic reduction in most pro-inflammatory
M1 markers (Tnfα, Mcp1, Itgax, Il6, and Nos2) with decreased
macrophage infiltration (F4/80 and Cd68) in MA-treated mice
3.5. MA Improves Obesity-Associated Metabolic Parameters in
WAT (Figure 5F). Furthermore, MA supplementation effectively
HFD-Fed Mice
alleviated hepatic steatosis and the expression of lipogenic genes
(Figure 5G). Collectively, these results indicate that MA was re-
Because MA (Figure 5A), a type of phenolic acid, was identified
sponsible for the metabolic effects of PE on HFD-induced obesity.
as the major antioxidant component present in PE in our pre-
vious studies,[9a,13b] we investigated whether MA was the active
constituent responsible for the anti-obesity effects of PE in mice. 4. Discussion
HFD-induced obese mice were orally administered with vehicle
(saline) or MA. A significant reduction in body weight during the Our previous studies of the chemical profiles and bioactive prop-
7 weeks of MA treatment was observed compared with the ve- erties of pear and recent clinical observations indicated that pear

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (6 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

Figure 4. PE reduces macrophage infiltration and pro-inflammatory phenotype in WAT. A) Macrophage-specific IBA1 immunostaining and number of
CLSs (asterisks) per high-power field (HPF) in WAT (10 HPF counted per mouse, n = 3 mice per group, magnification: ×200, scale bar: 50 µm). B)
mRNA levels for macrophage-specific and C) M1 and M2 type markers, and D) phosphorylation levels of JNK, ERK1/2, and p38 in WAT. Phosphorylation
of MAPKs was normalized to total MAPKs and conveyed as fold change compared to CD. Data are mean ± SEM; n = 5. *p < 0.05 versus CD; # p < 0.05
versus HFD.

can improve metabolic compilations in obesity.[8a,9a,9b,11] In the the ability to maintain and expand the pool of adipocytes in obe-
current study, we comprehensively assessed the therapeutic ef- sity, which requires further investigation.
fects of PE on HFD-induced obesity, IR, and hepatic steatosis In addition to the anti-adipogenic effect of PE, this extract also
and explored the underlying molecular mechanism. We found significantly downregulated HFD-induced lipogenic gene expres-
that PE treatment restored metabolic abnormalities in mice sion in both liver and WAT, although growing evidence suggests
chronically exposed to HFD. Five-week administration of PE re- that, unlike hepatic lipogenesis, increased WAT lipogenesis im-
sulted in significantly reduced body weight gain, fat mass, and proves metabolic phenotypes.[23] Consistent with our observa-
hyperlipidemia. Consistent with the results of a prior report tion, a number of studies have shown that HFD feeding leads
in streptozotocin-induced T2D mice,[13a] PE improved glucose to an increase in lipogenic gene expression in mice WAT.[24] Con-
homeostasis and insulin sensitivity as assessed by GTT, ITT, and sidering the diet composition used in our study and that used in
HOMA-IR. Interestingly, a recent meta-analysis of a prospec- these previous studies, it is possible that the results of our study
tive cohort also revealed an inverse association between apple are specific to comparison of the effects of HFD to those of a regu-
and pear consumption and T2D risk.[21] Thus, our findings sug- lar chow diet instead of to those of an ingredient-matched low-fat
gest that PE has therapeutic effects on T2D and cardiovascular diet.
disease, as obesity-associated IR is a major risk factor for these Numerous studies have demonstrated that hypertrophic ex-
diseases. pansion of adipocytes leads to dysfunctional adipose tissue asso-
Furthermore, the expansion of adipose tissue and hypertrophy ciated with increased infiltration and activation of inflammatory
of adipocytes in obese mice were markedly inhibited by PE. Lipid cells, lipolysis, and local and systemic IR both in humans and
accumulation and adipogenic genes were significantly reduced in animal models.[25] In the current study, PE attenuated the re-
in differentiated mature adipocytes and WAT, while UCP1 was cruitment of macrophages and increased the expression levels of
not enhanced in BAT and inguinal WAT, indicating that PE re- the pro-inflammatory M1 markers Tnfα, Mcp1, and Itgax in the
duced adiposity likely by inhibiting adipogenesis rather than via WAT of HFD-fed mice, indicating that PE supplementation nor-
thermogenic activation for energy expenditure. Although small malized the inflammatory phenotype of WAT. Intracellular sig-
adipocytes are more insulin sensitive in obesity, inhibiting adipo- naling pathways that regulate inflammation, such as the MAPK
genesis can lead to IR and ectopic liver fat accumulation because pathway, have been shown to regulate insulin sensitivity, obe-
of a reduced fat storage capacity.[22] However, we found that PE sity, and pathogenesis of diabetes.[26] For instance, JNK deficiency
treatment lowered hepatic fat accumulation and improved glu- attenuated obesity-induced IR by decreasing pro-inflammatory
cose homeostasis in HFD-fed mice, suggesting that PE improves macrophage polarization in mouse WAT,[20] and ERK1 knockout

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (7 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

Figure 5. MA improves HFD-induced obesity and associated metabolic complications in mice. Mice were fed a HFD for 9 weeks, followed by MA (40
mg kg−1 day−1 ) was orally administered for 7 weeks. A) Structure of MA. B) Body weight gain. C) GTT and D) ITT were performed at 15 weeks. E) mRNA
levels for adipogenic and adipokine genes and F) macrophage-specific and M1 type markers in WAT. G) H&E staining and mRNA levels for lipogenic
genes in liver (magnification: ×200, scale bar: 50 µm). Data are mean ± SEM; n = 5. Data are representative of two independent experiments with five
mice per group. *p < 0.05 versus CD; # p < 0.05 versus HFD.

mice were protected from HFD-induced obesity and IR because insulin sensitivity. Moreover, WAT inflammation in obese mice
of inhibited adipogenesis.[27] Although the role of p38 in obe- was significantly inhibited by MA, suggesting that MA is the anti-
sity in vivo remains unclear, a recent study showed that genetic obesity component present in pear fruits.
or pharmacological inhibition of p38α enhanced WAT brown- Taken together, our results demonstrate that PE and MA re-
ing, thereby preventing obesity in mice.[28] Here, we found that duce body weight gain, hepatic steatosis, WAT inflammation, and
PE treatment significantly reduced the hyperactivation of JNK, IR in HFD-induced obese mice and suggest the potential for di-
ERK1/2, and p38 MAPK in WAT of HFD-fed mice, suggesting etary supplementation with pear fruits to improve obesity and
that reduced adipogenesis, hypertrophy, and inflammation by PE obesity-related complications, including NAFLD and T2D. Fur-
are associated with deactivation of the MAPK signaling pathway ther studies are needed to determine the molecular targets of PE
in WAT. and its efficacy and safety in obese individuals.
We previously reported that MA is present mainly in pear fruits
(Pyrus pyrifolia)[12] and determined its antioxidative activity, ab-
sorption, and metabolism in vivo.[13b] However, no studies have
examined its anti-obesity effects. Although PE contains potent Supporting Information
antioxidants, such as chlorogenic acid and protocatechuic acid, Supporting Information is available from the Wiley Online Library or from
these compounds did not exert significant effects on 3T3-L1 dif- the author.
ferentiation (data not shown),[29] whereas MA markedly inhibited
adipogenesis and adipogenic gene expression levels. Thus, we
further analyzed the effects of MA on HFD-induced metabolic
dysregulation in mice. As observed during PE treatment, we Acknowledgments
found that 7-week administration of MA reversed HFD-induced X.T.T. performed most experiments, analyzed data, and co-wrote the
obesity and hepatic steatosis and improved glucose tolerance and manuscript. T.T.P.N. and C.H.J. performed in vitro experiments. M.J.K

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (8 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.mnf-journal.com

reviewed the manuscript and contributed to the discussion. S.L. and C.M [11] a) M. Conceicao de Oliveira, R. Sichieri, A. Sanchez Moura, Nutrition
performed histological analysis. J.H.M. performed PE and MA prepara- 2003, 19, 253; b) M. C. de Oliveira, R. Sichieri, R. Venturim Mozzer,
tion and contributed the discussion section. T.-I.J. designed the study and Appetite 2008, 51, 291.
co-wrote the manuscript. All authors read and reviewed the manuscript. [12] Y. G. Lee, J. Y. Cho, J. Park, S. H. Lee, W. S. Kim, K. H. Park, J. H. Moon,
This work was supported by High Value-Added Food Technology Devel- Food Sci. Biotechnol. 2013, 22, 1539.
opment Program through Korea Institute of Planning and Evaluation for [13] a) T. T. Wang, X. Li, B. Zhou, H. F. Li, J. Zeng, W. Y. Gao, J. Funct. Foods
Technology in Food, Agriculture, Forestry and Fisheries (IPET) funded by 2015, 13, 276; b) H. J. Lee, H. Y. Jeong, M. R. Jin, H. J. Lee, J. Y. Cho,
Ministry of Agriculture, Food and Rural Affairs (MAFRA) (315069-3) and J. H. Moon, Free Radic. Bio. Med. 2017, 110, 399.
NRF-2018R1D1A1B07041857.
[14] S. Reagan-Shaw, M. Nihal, N. Ahmad, FASEB J. 2008, 22, 659.
[15] T. M. Wallace, J. C. Levy, D. R. Matthews, Diabetes Care 2004, 27,
1487.
Conflicts of interest [16] a) A. M. Cypess, S. Lehman, G. Williams, I. Tal, D. Rodman, A. B.
Goldfine, F. C. Kuo, E. L. Palmer, Y. Tseng, A. Doria, G. M. Kolodny,
The authors declare no conflict of interest. C. R. Kahn, N. Engl. J. Med. 2009, 360, 1509; b) A. Bartelt, O. T. Bruns,
R. Reimer, H. Hohenberg, H. Ittrich, K. Peldschus, M. G. Kaul, U. I.
Tromsdorf, H. Weller, C. Waurisch, A. Eychmuller, P. L. S. M. Gordts,
F. Rinninger, K. Bruegelmann, B. Freund, P. Nielsen, M. Merkel, J.
Keywords Heeren, Nat. Med. 2011, 17, 200.
hepatic steatosis, inflammation, malaxinic acid, obesity, pears [17] L. A. Lotta, P. Gulati, F. R. Day, F. Payne, H. Ongen, M. van de Bunt,
K. J. Gaulton, J. D. Eicher, S. J. Sharp, J. N. Luan, E. D. Rolfe, I. D.
Stewart, E. Wheeler, S. M. Willems, C. Adams, H. Yaghootkar, N. G.
Received: December 10, 2018
Forouhi, K. T. Khaw, A. D. Johnson, R. K. Semple, T. Frayling, J. R. B.
Revised: March 30, 2019
Perry, E. Dermitzakis, M. I. McCarthy, I. Barroso, N. J. Wareham, D.
Published online: May 9, 2019
B. Savage, C. Langenberg, S. O’Rahilly, R. A. Scott, et al., Nat. Genet.
2017, 49, 317.
[18] a) C. N. Lumeng, J. L. Bodzin, A. R. Saltiel, J. Clin. Invest. 2007, 117,
[1] M. Ng, T. Fleming, M. Robinson, B. Thomson, N. Graetz, C. Mar- 175; b) A. Chawla, K. D. Nguyen, Y. P. S. Goh, Nat. Rev. Immunol.
gono, E. C. Mullany, S. Biryukov, C. Abbafati, S. F. Abera, J. P. Abra- 2011, 11, 738.
ham, N. M. Abu-Rmeileh, T. Achoki, F. S. AlBuhairan, Z. A. Alemu, [19] F. Bost, M. Aouadi, L. Caron, B. Binetruy, Biochimie 2005, 87, 51.
R. Alfonso, M. K. Ali, R. Ali, N. A. Guzman, W. Ammar, P. Anwari, A. [20] M. S. Han, D. Y. Jung, C. Morel, S. A. Lakhani, J. K. Kim, R. A. Flavell,
Banerjee, S. Barquera, S. Basu, D. A. Bennett, Z. Bhutta, J. Blore, N. R. J. Davis, Science 2013, 339, 218.
Cabral, I. C. Nonato, J. C. Chang, et al., Lancet 2014, 384, 766. [21] X. F. Guo, B. Yang, J. Tang, J. J. Jiang, D. Li, Food Funct. 2017, 8,
[2] V. S. Malik, W. C. Willett, F. B. Hu, Nat. Rev. Endocrinol. 2013, 9, 13. 927.
[3] N. Ouchi, J. L. Parker, J. J. Lugus, K. Walsh, Nat. Rev. Immunol. 2011, [22] B. Gustafson, S. Hedjazifar, S. Gogg, A. Hammarstedt, U. Smith,
11, 85. Trends Endocrinol. Metab. 2015, 26, 193.
[4] P. Arner, S. Bernard, M. Salehpour, G. Possnert, J. Liebl, P. Steier, B. [23] M. A. Herman, O. D. Peroni, J. Villoria, M. R. Schon, N. A. Abumrad,
A. Buchholz, M. Eriksson, E. Arner, H. Hauner, T. Skurk, M. Ryden, M. Bluher, S. Klein, B. B. Kahn, Nature 2012, 484, 333.
K. N. Frayn, K. L. Spalding, Nature 2011, 478, 110. [24] a) C. J. Chang, C. S. Lin, C. C. Lu, J. Martel, Y. F. Ko, D. M. Ojcius, S. F.
[5] a) S. P. Weisberg, D. McCann, M. Desai, M. Rosenbaum, R. L. Leibel, Tseng, T. R. Wu, Y. Y. M. Chen, J. D. Young, H. C. Lai, Nat. Commun.
A. W. Ferrante, Jr., J. Clin. Invest. 2003, 112, 1796; b) M. Kolak, J. 2015, 6, 7489; b) I. J. Lodhi, L. Yin, A. P. L. Jensen-Urstad, K. Funai, T.
Westerbacka, V. R. Velagapudi, D. Wagsater, L. Yetukuri, J. Makkonen, Coleman, J. H. Baird, M. K. El Ramahi, B. Razani, H. W. Song, F. H.
A. Rissanen, A. M. Hakkinen, M. Lindell, R. Bergholm, A. Hamsten, Fong, J. Turk, C. F. Semenkovich, Cell Metab. 2012, 16, 189; c) Y. Qiao,
P. Eriksson, R. M. Fisher, M. Oresic, H. Yki-Jarvinen, Diabetes 2007, J. Sun, S. F. Xia, X. Tang, Y. H. Shi, G. W. Le, Food Funct. 2014, 5, 1241;
56, 1960. d) Y. S. Lee, S. H. Kim, H. J. Yuk, G. J. Lee, D. S. Kim, Nutrients 2018,
[6] D. Ryan, M. Heaner, Obesity 2014, 22 Suppl 22, S1. 10, E1087; e) T. Li, J. Gao, M. Du, J. Song, X. Mao, Nutrients 2018, 10,
[7] a) L. A. Bazzano, M. K. Serdula, S. Liu, Curr. Atheroscler. Rep. 2003, 5, E331.
492; b) B. J. Rolls, J. A. Ello-Martin, B. C. Tohill, Nutr. Rev. 2004, 62, 1. [25] a) B. Gustafson, S. Gogg, S. Hedjazifar, L. Jenndahl, A. Hammarst-
[8] a) J. Y. Cho, S. H. Lee, E. H. Kim, H. R. Yun, H. Y. Jeong, Y. G. Lee, W. edt, U. Smith, Am. J. Physiol. Endocrinol. Metab. 2009, 297, E999; b)
S. Kim, J. H. Moon, Biosci., Biotechnol. Biochem. 2015, 79, 260; b) Y. I. Wernstedt Asterholm, C. Tao, T. S. Morley, Q. A. Wang, F. Delgado-
G. Lee, J. Y. Cho, C. M. Kim, S. H. Lee, W. S. Kim, T. I. Jeon, K. H. Park, Lopez, Z. V. Wang, P. E. Scherer, Cell Metab. 2014, 20, 103; c) L. A.
J. H. Moon, Food Sci. Biotechnol.2013, 22, 803. Muir, N. A. Baker, A. R. Washabaugh, C. K. Neeley, C. G. Flesher, J.
[9] a) K. H. Lee, J. Y. Cho, H. J. Lee, K. Y. Park, Y. K. Ma, S. H. Lee, J. A. B. DelProposto, L. M. Geletka, A. A. Ghaferi, J. F. Finks, K. Singer, O.
Cho, W. S. Kim, K. H. Park, J. H. Moon, Food Sci. Biotechnol. 2011, 20, A. Varban, C. N. Lumeng, R. W. O’Rourke, Adipocyte 2017, 6, 134.
1539; b) K. H. Lee, J. Y. Cho, H. J. Lee, Y. K. Ma, J. Kwon, S. H. Park, [26] H. Gehart, S. Kumpf, A. Ittner, R. Ricci, EMBO Rep. 2010, 11, 834.
S. H. Lee, J. A. Cho, W. S. Kim, K. H. Park, J. H. Moon, J. Agric. Food [27] F. Bost, M. Aouadi, L. Caron, P. Even, N. Belmonte, M. Prot, C. Dani,
Chem. 2011, 59, 10124; c) X. Li, W. Y. Gao, L. J. Huang, J. Y. Zhang, P. Hofman, G. Pages, J. Pouyssegur, Y. Le Marchand-Brustel, B. Bi-
X. H. Guo, J. Food Sci. 2011, 76, C985; d) X. Li, J. Y. Zhang, W. Y. Gao, netruy, Diabetes 2005, 54, 402.
Y. Wang, H. Y. Wang, J. G. Cao, L. Q. Huang, J. Agric. Food Chem. [28] S. Zhang, H. Cao, Y. Li, Y. Jing, S. Liu, C. Ye, H. Wang, S. Yu, C. Peng,
2012, 60, 8738; e) M. Y. Heng, S. Katayama, T. Mitani, E. S. Ong, S. L. Hui, Y. C. Wang, H. Zhang, F. Guo, Q. Zhai, H. Wang, R. Huang, L.
Nakamura, Food Chem. 2017, 221, 1388. Zhang, J. Jiang, W. Liu, H. Ying, PLoS Biol. 2018, 16, e2004225.
[10] S. P. Sharma, H. J. Chung, H. J. Kim, S. T. Hong, Nutrients 633, 2016, [29] C. Maki, M. Funakoshi-Tago, R. Aoyagi, F. Ueda, M. Kimura, K. Ko-
8. bata, K. Tago, H. Tamura, PLoS One 2017, 12, e0173264.

Mol. Nutr. Food Res. 2019, 63, 1801347 1801347 (9 of 9) 


C 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
Copyright of Molecular Nutrition & Food Research is the property of John Wiley & Sons,
Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv
without the copyright holder's express written permission. However, users may print,
download, or email articles for individual use.

You might also like