Open Access

Anti‑obesity effect in high‑fat‑diet‑induced obese C57BL/6 mice: Study of a novel extract from mulberry (Morus alba) leaves fermented with Cordyceps militaris

  • Authors:
    • Mi Rim Lee
    • Ji Eun Kim
    • Jun Young Choi
    • Jin Ju Park
    • Hye Ryeong Kim
    • Bo Ram Song
    • Young Whan Choi
    • Kyung Mi Kim
    • Hyunkeun Song
    • Dae Youn Hwang
  • View Affiliations

  • Published online on: January 21, 2019     https://doi.org/10.3892/etm.2019.7191
  • Pages: 2185-2193
  • Copyright: © Lee et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The therapeutic effects of mulberry (Morus alba) leaves on lipid metabolism, including lipogenesis, lipolysis and hyperlipidemia are widely known, although their fermented products are yet to be applied. To investigate the therapeutic effects of a novel extract of mulberry leaves fermented with Cordyceps militaris (EMfC) on lipid metabolism, the lipid profile of serum, lipid accumulation, lipolytic activity and lipogenesis regulation were measured in high fat diet (HFD)‑induced obese C57BL/6 mice treated with EMfC for 12 weeks. Briefly, the concentrations of low‑density lipoprotein, triglyceride, total cholesterol and glucose significantly decreased in the serum of the HFD+EMfC treated group when compared with the HFD+Vehicle treated group, while the levels of high‑density lipoprotein increased in the HFD+EMfC group. The amount of abdominal fat and the size of adipocytes were significantly lower in the HFD+EMfC treated group when compared with the HFD+Vehicle treated group. The weight and number of lipid droplets of liver tissue exhibited a similar decrease. Furthermore, the mRNA levels of peroxisome proliferator‑activated receptor‑γ for adipogenesis as well as adipocyte protein 2 and Fas cell surface death receptor for lipogenesis reduced following EMfC treatment for 12 weeks. Phosphorylation of perilipin and hormone‑sensitive lipase, and in the adipose triglyceride lipase expression showed a significant increase in the HFD+EMfC treated group. These results indicated that EMfC may prevent fat accumulation in the HFD‑induced obese C57BL/6 mice through the inhibition of lipogenesis and by stimulating lipolysis. Thus, the results provide evidence for the potential use of EMfC as an anti‑obesity complex in the treatment of obesity.

Introduction

Globally, obesity is a serious health problem due to its strong association with increased dyslipidemia, cardiovascular disease (including hypertension, stroke, myocardial infarction), insulin resistance, glucose metabolism disorders, osteoarthritis and some cancers (1,2). According to a recent report, the increase in the world obesity rate from 1975 to 2014 was 3.2–10.8% in men, and 6.4–14.4% in women (3). This trend will have a significant impact on world health and the economy. The main causes of obesity are the delicious and energetic diets rich in fat, consumption imbalances, low momentum, fat, sugar and salt (1). The most common methods for preventing and treating obesity are maintaining an optimal weight by calibrating lifestyle habits by introducing well-structured calorie restriction and exercise programs (4). Lifestyle corrections are very challenging for obese patients, who subsequently resort to the use of medications as an adjunct. However, factors such as the cost and potential side effects are the main causes for failure of weight and obesity management with medication. Recent reports indicate that many herbal extracts, including Paullinia cupana, Plantago psyllium, Garcinia cambogia, and Ilex paraguariensis, are relatively safe for controlling weight, by promoting the lipid and carbohydrate metabolism (5). Thus, herbal extracts from natural sources are likely to be safe and effective alternatives for treating obesity.

Commonly known as mulberry in Korea, Morus alba (family: Mulberry), grows in northern China, and has been cultivated from India to the Middle East, southern Europe, and recently North America. Pharmacological studies have reported the roots and bark to have antibacterial (6), antioxidant and hypoglycemic (7,8), and neuroprotective, anti-ulcer, analgesic and anti-inflammatory activities (912). Several studies have also presented the beneficial effects of mulberry leaves on lipid metabolism. Kobayashi et al (13) reported that administration of mulberry leaves to rats for 7 weeks induced the upregulation of fatty acid oxidation and downregulation of lipogenesis-related genes. Other studies reported that the mulberry leaf extract significantly reduced the lipid accumulation and lipoprotein-related protein expression in 3T3-L1 adipocytes (14). Additionally, the mulberry leaf extract stimulates the glucose uptake in rat adipocytes, and reduces the adipocyte development in white adipose tissue extracted from db/db mice through inhibition of oxidative stress (1517). Cordyceps militaris (C. militaris) is an insect pathogen belonging to Ascomycota, and is traditionally used in Chinese and East Asian medicine (18,19). Cordycepin isolated from C. militaris is effective in anticancer (20), antitumor (21), antimicrobial (22) and antioxidant (23) therapy. Recent reports indicate that C. militaris-fermented products are beneficial in attenuating liver fibrosis (24,25); similar effects were observed with a mixture of C. militaris and a secondary metabolite secreted from mushroom (26). In our previous study, we reported that the extract of Morus alba leaves fermented with C. militaris (EMfC) has a lipolytic effect on adipocytes isolated from SD rats (27). However, no detailed study was performed to evaluate the inhibitory effect of EMfC on obesity.

Based on previous reports, we hypothesized that the EMfC induces its anti-obesity effects through the regulation of lipid metabolism. We therefore used mice with high-fat diet-induced obesity to evaluate changes in fat accumulation and lipid profiles, as well as examined the underlying mechanisms of adipogenesis, lipogenesis and lipolysis.

Materials and methods

Preparation of EMfCs

One of the authors (Professor Young Whan Choi) characterized the samples of mulberry leaves collected from plantations in the Sangju district of Korea in October 2015. Voucher specimens (accession no. Mul-PDRL-1) were deposited in the herbarium of the Pusan National University (Miryang, Korea). Professor Sang Mong Lee of the Department of Life Science and Environmental Biochemistry, Pusan National University, kindly provided the C. militaris used for fermentation. The Jeongeup Agriculture Cooperative Federations for Silkworm Farming (Jeongeup, Korea) supplied the silkworm pupae powder.

Briefly, the mulberry leaves were powder using an electric blender, after drying in a hot-air drying machine (JSR, Seoul, Korea) for 24 h at 60°C. The mulberry powder was sterilized by autoclaving at 121°C for 60 min, mixed with 50% silkworm powder (SWP), and the mixture inoculated with 10% C. military (v/w). The samples were incubated in a shaking incubator (cat. no. SI-600R; Lab Companion, Seoul, Korea) at 150 rpm and 25°C, and fermented for 4 weeks. On the last day of fermentation, the mixture was harvested from the flask and extracted with 95% EtOH. To prepare the extracts of fermented mulberry powder, the extracted powder was mixed with the solvent (95% EtOH) in a fixed liquid ratio (mulberry powder:solvent, ratio 1:10) and sonicated for 1 h using a JAC ultrasonic device (KODO, Hwangseong, Korea). Centrifuging at 3,000 rpm for 10 min separated the supernatant from the sonicated extract. The pellet was resuspended in 9 ml of the solvent, and further sonicated using the same conditions. This procedure was repeated once more, and the resultant supernatant was collected, filtered through a 0.4 µm filter (cat. no. HAWP04700; Millex-LH, MerckMillipore, Darmstadt, Germany), and evaporated using a vacuum evaporator (cat. no. R-300, BUCHI Corporation, New Castle, Delaware, USA). Finally, lyophilization of the EMfC was achieved using the circulating extraction equipment (IKA Labortechnik, Staufen, Germany). The extracts were dissolved in DMSO (cat. no. D2660, Sigma-Aldrich Co., St Louis, MO, USA) at a concentration of 50 mg/ml before use.

Design of animal experiment

Eight-week-old C57BL/6 male mice were purchased from Samtako Bio-Korea Inc. (Osan, Korea), and provided ad libitum access to water and a standard irradiated chow diet (Samtako Bio-Korea Inc.). During the experiment, mice were maintained in a pathogen-free state under a strict light cycle (lights on at 08:00 h and off at 20:00 h) at 23±2°C and 50±10% relative humidity. All C57BL/6 mice were handled at the Pusan National University-Laboratory Animal Resources Center, accredited by the Korea Food and Drug Administration (FDA; Accredited Unit no. 000231) and AAALAC International (Accredited Unit no. 001525).

The Pusan National University Animal Ethics Committee (PNU-2017-1519) approved the protocol for animal study. All animals were acclimatized on a normal diet (D12450K; Research Diets, New Brunswick, NJ, USA) for 1 week. The C57BL/6 mice were then divided into 4 study groups (7 mice/group): (1) Control group fed a normal diet (No treatment group), (2) group fed HFD (high fat diet) plus vehicle (olive oil+1% DMSO) (HFD+Vehicle treated group), (3) group fed HFD plus 10 mg/kg Orlistat (Sigma-Aldrich Co.) (HFD+OT treated group), and (4) group fed HFD plus 50 mg/kg EMfC (HFD+EMfC treated group). For 12 weeks, mice of all HFD treatment groups consumed HFD containing 60% kcal fat purchased from Research Diets (cat. no. D12492; Research Diets, Inc., New Brunswick, USA). After 24 h of the final EMfC treatment, all mice were euthanized using CO2 gas, after which the tissue samples were acquired and stored in Eppendorf tubes at −70°C until assay.

Measurement of body weight, dietary intake and organ weight

Throughout the experimental period, the body weight of mice treated with Vehicle, OT or EMfC were measured daily at 10:00 am using an electronic balance (cat. no. AD-2.5; Mettler Toledo, Greifensee, Switzerland) according to the KFDA guidelines. In addition, the weights of liver and abdominal organs collected from the sacrificed C57BL/6 mice were determined using the same method employed to measure the body weight. Feed intake was measured at the same time once a week during the study period, using a chemical balance (Mettler Toledo, Switzerland).

Serum biochemical analysis

After 12 weeks of feeding experimental diets, blood was collected from the abdominal veins of all C57BL/6 mice after fasting for 18 h. The blood samples were incubated for 30 min at room temperature in serum separating tubes (BD Biosciences, Franklin Lakes, NJ, USA). Serum was obtained by centrifugation at 1,500 × g for 15 min. Serum glucose, TC (total cholesterol), TG (triglyceride), high-density lipoprotein (HDL)-cholesterol, and low-density lipoprotein (LDL)-cholesterol levels were analyzed by the automatic chemical analyzer (BS-120 Chemistry Analyzer; Mindray, Shenzhen China). Additionally, serum was analyzed for alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), and creatinine (Crea) by using an automatic biochemical analyzer (BS-120; Mindray). All assays were conducted in duplicate using fresh serum.

Histopathological analysis

Liver and fat tissues dissected from mice of all subset groups were fixed in 10% neutral buffered formaldehyde (pH 6.8) overnight. The dehydrated liver tissue was then embedded in paraffin wax. Next, a series of liver and fat sections (4 µm) were cut from the paraffin-embedded tissues using a Leica microtome (cat. no. DM500; Leica Microsystems, Bannockburn, IL, USA). These sections were then deparaffinized with xylene (cat. no. 8587-4410; Daejung, Gyeonggi-do, Korea), rehydrated with graded ethanol (decreasing concentrations of 100-70%), and finally washed with distilled water. The slides with liver sections were stained with hematoxylin (cat. no. MHS16) and eosin (cat. no. HT110332; both Sigma-Aldrich Co.), washed with dH2O, and pathological changes were assessed using the Leica Application Suite (Leica Microsystems).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Frozen liver tissue was chopped with scissors and homogenized in RNA Bee solution (cat. no. CS-105B; Tet-Test Inc., Friendswood, TX, USA). Total RNA molecules were isolated by centrifugation at 15,000 rpm for 15 min, after which the concentration was measured by UV spectroscopy. The complementary DNA (cDNA) was synthesized by Invitrogen Superscript II reverse transcriptase (cat. no. 4376600; Thermo Scientific, Wilmington, Delaware, USA). Quantitative PCR was performed with the cDNA template (2 µl) and 2X Power SYBR-Green (6 µl; Toyobo Life Science, Osaka, Japan) containing specific primers. The primer sequences for target gene expression identification used were as follows: PPARγ sense, 5′-GAGTTCATGCTTGTGAAGGATGCAAGG-3′ and anti-sense, 5′-CATACTCTGTGATCTCTTGCACG-3′; C/EBPα sense, 5′-GTGGACAAGAACAGCAACGAGTACC-3′ and anti-sense, 5′-GGAATCTCCTAGTCCTGGCTTGC-3′; β-actin sense, 5′-TGGAATCCTGTGGCATCCATGAAAC-3′ and anti-sense, 5′-TAAAACGCAGCTCAGTAACAGTCCG-3′. qPCR was performed for 40 cycles using the following parameters: Denaturation at 95°C for 15 sec, followed by annealing and extension at 70°C for 60 sec. Fluorescence intensity was measured at the end of the extension phase of each cycle. The threshold value for the fluorescence intensities of all samples was set manually. The reaction cycle at which the PCR products exceeded this fluorescence intensity threshold during the exponential phase of PCR amplification was considered as be the threshold cycle (Cq). Expression of the target gene was quantified relative to that of the housekeeping genes β-actin, based on comparison of the Cqs at a constant fluorescence intensity, as per Livak and Schmittgen's method (28).

Western blot analysis

Liver tissue (50 mg) collected from each group was homogenized using PRO-PREPTM Solution (cat. no. 170841; iNtRON Biotechnology Inc., Sungnam, Korea), and total protein extracts were collected by centrifugation at 13,000 rpm for 5 min. The prepared proteins were subsequently subjected to 10% SDS-PAGE for 2 h at 100 V, and transferred to a nitrocellulose membrane (GE Healthcare, Little Chalfont, UK) for 2 h at 40 V in transfer buffer (25 mM Trizma-base, 192 mM glycine, and 20% methanol). Membranes were then exposed to appropriate dilutions of primary antibodies and allowed to hybridize overnight at 4°C: anti-perilipin antibody (cat. no. 9349S), anti-p-perilipin antibody (cat. no. 9621), anti-HSL antibody (cat. no. 4107S), anti-p-HSL antibody (cat. no. 4139S), anti-ATGL antibody (cat. no. 2183S) and anti-β-actin antibodies (cat. no. 4967S; all Cell Signaling Technology, Danvers, MA, USA). After removal of the antibodies, the membranes were washed three times in a 10 mM Trizma-base (150 mM NaCl and 0.05% Tween-20) solution for 10 min. The membranes were subsequently incubated with horseradish peroxidase-conjugated anti-secondary antibody for 1 h at room temperature, after which they were washed again as described above, and developed using the Enhanced Chemiluminescence Reagent Plus kit (cat. no. DG-WF100; Dogen, Seoul, Korea). Finally, the results were quantified using the Image Analyzer System (Fluorchem FC2; Alpha Innotech, CA, USA) and expressed as the fold-increase over control values.

Statistical significance analysis

One-way analysis of variance was performed using SPSS 10.10 (SPSS, Inc., Chicago, IL, USA) in order to determine the variance and identify significant differences between the treated group and other groups, as well as between the vehicle group and other HFD fed groups. All values are presented as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of EMfC on body weight and lipid profile of serum

To evaluate the anti-obesity effects of EMfC, we analyzed the changes in body weight and serum lipid profile of HFD-induced obesity mice treated with EMfC for 12 weeks (Fig. 1). Although no significant differences were detected in the body weight of HFD+Vehicle treated group and HFD+EMfC treated group, the serum lipid profile differed after EMfC treatment. As shown in Fig. 1B, all lipid profile factors were 40–180% higher in the HFD+Vehicle treated group than in the No treatment group. However, the concentration of LDL, TG, TC and glucose significantly decreased in the HFD+EMfC treated group as compared to the HFD+Vehicle treated group (P<0.005; Fig. 1B). Additionally, EMfC treatment resulted in enhanced HDL levels (Fig. 1B). These results suggest that EMfC induces a recovery of the serum lipid profile in HFD-induced obesity mice, although it is unable to inhibit weight gain due to HFD.

Effect of EMfC on abdominal fat accumulation

In order to investigate the effect of EMfC on abdominal fat accumulation, we assessed the amount of abdominal fat and the size of adipocytes by H&E staining. The HFD+Vehicle treated group had higher content of abdominal fat as compared to the No treatment group (P<0.005). However, levels significantly decreased in the HFD+EMfC and HFD+OT treated groups as compared to the HFD+Vehicle treated group (P<0.005; Fig. 2A). A similar pattern was observed for the average area of single adipocyte of fat tissue. HFD+EMfC and HFD+OT treated groups showed significant decrease of the adipocyte area as compared with the HFD+Vehicle treated group (P<0.005; Fig. 2B). These results suggest that EMfC prevents the accumulation of abdominal fat in HFD-induced obesity mice.

Effects of EMfC on hepatic steatosis of HFD-induced obesity mice

Liver weight alterations and hepatic steatosis are common obesity-related phenomena. Therefore, we measured the changes in weight and fat accumulation of liver in HFD-induced obesity mice treated with EMfC. The HFD+Vehicle treated group showed increased liver weight compared to the No treatment group (P<0.005). These changes recovered with OT or EMfC treatment (Fig. 3A). Also, significant changes were detected in the number of lipid droplet in the liver tissues of all groups stained with H&E. The liver section of the HFD+Vehicle treated group showed dramatic increase in the number of lipid drops compared to the No treatment group. In comparison, only a few lipid drops were seen in the HFD+OT and HFD+EMfC treated groups (P<0.005; Fig. 3B). These results indicate that EMfC significantly inhibits the liver hypertrophy, fat accumulation and hepatic steatosis in HFD-induced obesity mice.

Effects of EMfC on adipogenesis and lipogenesis of HFD-induced obese mice

To further elucidate the mechanisms by which EMfC affects obesity, we measured the mRNA levels of several key genes that affecting adipogenesis and lipogenesis in the liver tissue. As shown in Fig. 4, there were significant changes in the mRNA levels of adipogenesis and lipogenesis related genes after EMfC treatment. The mRNA levels of PPARγ and C/EBPα for adipogenesis increased after HFD feeding (HFD+Vehicle treated group) compared to the No treatment group (P<0.005). However, these levels were dramatically decreased after OT or EMfC treatment (HFD+OT and HFD+EMfC treated groups, respectively), although no changes were seen in the C/EBPα level (Fig. 4A). The gene expression of aP2 and FAS were showed dramatically increased in the HFD+Vehicle treated group, but significantly decreased in the HFD+OT and HFD+EMfC treated groups when compared to the No treatment group (P<0.005; Fig. 4B). These results indicate that EMfC treatment probably inhibits adipogenesis and lipogenesis in the liver tissue through the regulation of PPARγ, aP2 and FAS expression.

Effect of EMfC on lipolysis of HFD-induced obese mice

We finally investigated the molecular mechanism for lipolytic activity of EMfC by analyzing the expression and activation of key enzymes involved in triglyceride metabolism. We evaluated the phosphorylation or expression of three major enzymes, including perilipin, HSL and ATGL. We observed increased protein levels of ATGL in the liver tissues after treatment in the HFD+OT and HFD+EMfC groups, as compare to No treatment and HFD+Vehicle treated groups (P<0.005; Fig. 5). The protein levels of perilipin and HSL significantly decreased after OT and EMfC treatment, compared to the HFD+Vehicle treated group. Interestingly, the phosphorylation of perilipin and HSL were significantly enhanced in the HFD+EMfC treated group, while an increase in phosphorylation of perilipin was observed in the HFD+OT treated group (P<0.005; Fig. 5). Our data indicates that increased lipolytic ability of EMfC is mainly dependent on increased expression of ATGL and increased activation of perilipin, as well as increased activation of HSL, unlike OT. We therefore suggest that compared to OT, EMfC is likely to be more efficient in contributing to lipolysis.

Discussion

Recent pharmacological approaches for treating and controlling obesity have resorted to the use of drugs to induce anorexia, inhibit nutritional absorption, and promote weight loss (2). Clinically available anti-obesity agents include pancreatic lipase inhibitors such as orlistat, and appetite suppressants that act on the central nervous system, such as lorcaserin (29,30). Since body fat accumulation is a major risk factor for chronic diseases including hyperlipidemia, diabetes, cardiovascular disease and cancer, interest has increased for developing of new drugs having lipolytic and lipogenic activities (31,32). Numerous studies reported that several lipolytic agents, including norepinephrine, isoproterenol, phoscholine, dibutyl-cAMP (DBcAMP) and theophylline induce lipolysis in adipocytes (33). However, these anti-obesity drugs are associated with serious side effects such as constipation, insomnia, vomiting, headache, abdominal pain, and myocardial infarction (34). Therefore, the development of anti-obesity agents derived from natural products that are effective in weight management in suppressing and decomposing the fat accumulation, and having minimal side effects, are required (35). In this study, to analyze the anti-obesity effect of EMfC, we used orlistat as a positive control for assessing the anti-obesity effect in HFD-induced obese mice. At the end of the 12 weeks study period, although the HFD treated mice had maximum weight gain, there was no effective weight loss with orlistat and EMfC treatments also. This could be because orlistat is an anti-obesity agent involved in fat metabolism rather than weight loss effect. Secondly, the concentration of orlistat used in the study is 10 mg/kg, which is a low concentration as compared to the commonly used concentration of 10–50 mg/kg. The third reason is that although the final effect of orlistat is reduction of body weight following the suppression of fat absorption, 12 weeks is considered the period when no effective weight loss has occurred. Subsequently, we expect similar effects of orlistat and EMfC on fat metabolism, resulting in weight loss during longer study protocols.

In addition, to study the changes in body weight as a consequence of changes in dietary intake (data not shown), a slight decrease was observed in dietary intake of the experimental group (including the vehicle group), with no obvious difference between. We believe this slightly reduction in consumption may be due to the vehicle, and not olistat and EMfC.

Our study data reveals that EMfC treatment inhibits the visceral (epididymal and abdominal) adipose tissue mass, adipocyte cell size, serum LDL, serum TG, serum TC and serum glucose in obese mice induced by HFD. In addition, decreased liver weight and decreased lipid drop after EMfC exposure suggests that EMfC has an anti-obesity effect by being involved in liver lipid metabolism. Interestingly, the serum analysis of the experimental group including EMfC treatment for 12 weeks showed no adverse side effects such as hepatotoxicity and kidney toxicity (data not shown). Taken together these results suggest that the reduction in fat mass observed in HFD+EMfC treated mice is due not to the toxicity of EMfC, but due to the direct pharmacological action on adipose tissue.

EMfC treatment in HFD fed mice decreased the serum TG, TC, LDL and glucose levels proportionate to the decrease in adipose tissue mass. Conversely, the serum HDL concentration dramatically increased after EMfC and OT treatment. triglycerides (TGs) accumulate in adipose tissues by forming lipid drops of adipocytes (36). Our data displayed that the level of TG only marginally after EMfC treatment. Microscopic and histologic examinations revealed reduction in the mass of adipose tissue and adipocyte cell sizes of the HFD+EMfC treated groups as compared to the HFD+Vehicle treated group. The liver weight and number of lipid drops also decreased in the HFD+EMfC treated organ. Although the effect on serum level reduction of accumulated TGs is marginal, EMfC reduces adipose tissue due to decreasing of adipocyte size and cell number, and it is therefore possible to control cardiovascular diseases by decreasing the concentration of LDL and TC in serum. Therefore, we believe that the association between cytokine and hormones involved in fat accumulation (such as adipokine and leptin), and the relationship with cardiovascular diseases should be studied further, with respect to the anti-obesity effects of EMfC.

Adipose tissue is responsible for systemic energy homeostasis. PPARγ, a predominant transcription factor in adipose tissue, plays an important role in adipocyte differentiation, lipid storage, and glucose homeostasis (37,38). PPARγ regulates the gene expression of enzymes involved in the lipogenesis of adipose tissue, such as FAS, aP2, and lipoprotein lipase (39,40). FAS is a major lipogenic enzyme that catalyzes the biosynthesis of long chain fatty acids from acetyl-CoA precursors (41), and aP2 is an intracellular fatty acid binding protein that binds long-chain fatty acids (42). In this study, EMfC significantly reduced the mRNA levels of PPARγ, aP2, and FAS in the HFD-induced obesity mice. These results suggest that EMfC treatment improves the intracellular fatty acid metabolism by inhibiting the lipogenic gene expression. Interestingly, there was no inhibition in the expression of C/EBPα, a transcription factor similar to PPARγ and involved in lipogenesis, after EMfC administration. Orlistat inhibits the expression of two transcription factors (PPARγ and C/EBPα) involved in lipid synthesis, suggesting that EMfC has a different mechanism for inhibiting lipid synthesis.

In addition to the synthesis of TG stored in adipose tissue, lipolysis (hydrolysis) is an important mechanism for fat accumulation in adipose tissue. ATGL (PNPLA2, desnutrin) is an important enzyme that initiates TG degradation and produces diacylglycerol (DG). It serves to initiate the sequential process of TG digestion, limiting the rate at the first step and determining the maximum rate for fatty acid mobilization (4345). We observed that EMfC increases the amount of ATGL protein expression, similar to orlistat treatment. Perilipin is activated by the phosphorylation by phosphorylated PKA, which in turn activates the lipase of ATGL (46). Perilipin phosphorylation also increases after exposure to orlistat or EMfC, suggesting that EMfC has a similar lipid-breakdown mechanism to the orlistat induced mechanism. Interestingly, the phosphorylation of HSL, where ATGL degrades the DG into monoglycerides, occurs more strongly in EMfC than in orlistat (47), suggesting that EMfC has better activity in lipolysis than orlistat.

In conclusion, our study demonstrates that EMfC treatment successfully reduces the accumulation of adipose tissue and adipocyte cell size, and decrease the serum LDL, TG, TC and glucose levels in HFD-induced obese mice. In addition, there is a decrease in the liver weight and lipid drops, suppression in the expression of the lipogenic gene, and induction in the expression and activation of lipolysis protein. The strong effects of EMfC on fat loss and fatty liver loss without any accompanying adverse health effects suggests EMfC to be a potential anti-obesity drug that results in lipolysis.

Acknowledgements

The abstract was presented at the Korean Association for Laboratory Animal Science Winter Symposium Jan 24-Jan 27 2018 in Wonju, Kanwon-do, Korea and published as abstract no. OR-S-06 in Lab Anim Res 34 (Suppl. 1): 2018. The authors would like to thank their animal technician, Miss. Jin Hyang Hwang, for directing the animal care at the Laboratory Animal Resources Center in Pusan National University.

Funding

The present study was supported by grants from the Korea Institute of Planning & Evaluation for Technology of Food, Agriculture and Forestry (grant no. 116027-032-HD030).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

MRL, JEK, JYC, JJP, HRK, BRS, YWC, KMK and DYH participated in the design of the study, sample preparation, animal experiments and data analyses. HS performed the data analysis and assisted with manuscript preparation.

Ethics approval and consent to participate

The Pusan National University Animal Ethics Committee (no. PNU-2017-1519) approved the protocol for the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EMfC

extract of mulberry leaves fermented with Cordyceps militaris

HFD

high fat diet

TG

triglyceride

TC

total cholesterol

References

1 

Haslam DW and James WP: Obesity. Lancet. 366:1197–1209. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Després JP and Lemieux I: Abdominal obesity and metabolic syndrome. Nature. 444:881–887. 2006. View Article : Google Scholar : PubMed/NCBI

3 

NCD Risk Factor Collaboration (NCD-RisC): Worldwide trends in diabetes since 1980: A pooled analysis of 751 population-based studies with 4.4 million participants. Lancet. 387:1513–1530. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Maguire T and Haslam D: The Obesity Epidemic and its Management. Matthew Wright: Pharmaceutical Press; London: pp. 264–267. 2010

5 

Pittler MH, Schmidt K and Ernst E: Adverse events of herbal food supplements for body weight reduction: Systematic review. Obes Rev. 6:93–111. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Gunjal S, Ankola AV and Bhat K: In vitro antibacterial activity of ethanolic extract of Morus alba leaf against periodontal pathogens. Indian J Dent Res. 26:533–536. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Raman ST, Ganeshan AK, Chen C, Jin C, Li SH, Chen HJ and Gui Z: In vitro in vivo antioxidant activity of flavonoid extracted from mulberry fruit (Morus alba L.). Pharmacogn Mag. 12:128–133. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Wang Y, Xiang L, Wang C, Tang C and He X: Antidiabetic and antioxidant effects and phytochemicals of mulberry fruit (Morus alba L.) polyphenol enhanced extract. PLoS One. 8:e711442008. View Article : Google Scholar

9 

Jo SP, Kim JK and Lim YH: Antihyperlipidemic effects of stilbenoids isolated from Morus alba in rats fed a high-cholesterol diet. Food Chem Toxicol. 65:213–218. 2014. View Article : Google Scholar : PubMed/NCBI

10 

El-Beshbishy HA, Singab AN, Sinkkonen J and Pihlaja K: Hypolipidemic and antioxidant effects of Morus alba L. (Egyptian mulberry) root bark fractions supplementation in cholesterol-fed rats. Life Sci. 78:2724–2733. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Eo HJ, Park JH, Park GH, Lee MH, Lee JR, Koo JS and Jeong JB: Anti-inflammatory and anti-cancer activity of mulberry (Morus alba L.) root bark. BMC Complement Altern Med. 14:2002014. View Article : Google Scholar : PubMed/NCBI

12 

Chan EW, Lye PY and Wong SK: Phytochemistry, pharmacology, and clinical trials of Morus alba. Chin J Nat Med. 14:17–30. 2016.PubMed/NCBI

13 

Kobayashi Y, Miyazawa M, Kamei A, Abe K and Kojima T: Ameliorative effects of mulberry (Morus alba L.) leaves on hyperlipidemia in rats fed a high-fat diet: Induction of fatty acid oxidation, inhibition of lipogenesis, and suppression of oxidative stress. Biosci Biotechnol Biochem. 74:2385–2395. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Yang SJ, Park NY and Lim Y: Anti-adipogenic effect of mulberry leaf ethanol extract in 3T3-L1 adipocytes. Nutr Res Pract. 8:613–617. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Ann JY, Eo HY and Lim YS: Mulberry leaves (Morus alba L.) ameliorate obesity-induced hepatic lipogenesis, fibrosis, and oxidative stress in high-fat diet-fed mice. Genes Nutr. 10:462015. View Article : Google Scholar : PubMed/NCBI

16 

Naowaboot J, Pannangpetch P, Kukongviriyapan V, Prawan A, Kukongviriyapan U and Itharat A: Mulberry leaf extract stimulates glucose uptake and GLUT4 translocation in rat adipocytes. Am J Chin Med. 40:163–175. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Sugimoto M, Arai H, Taura Y, Murayama T, Khaengkhan P, Nishio T, Ono K, Ariyasu H, Akamizu T, Ueda Y, et al: Mulberry leaf ameliorates the expression profile of adipocytokines by inhibiting oxidative stress in white adipose tissue in db/db mice. Atherosclerosis. 204:388–394. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Sung GH, Hywel-Jones NL, Sung JM, Luangsa-Ard JJ, Shrestha B and Spatafora JW: Phylogenetic classification of Cordyceps and the clavicipitaceous fungi. Stud Mycol. 57:5–59. 2007. View Article : Google Scholar : PubMed/NCBI

19 

De Silva DD, Rapior S, Sudarman E, Stadler M, Xu J, Alias SA and Hyde KD: Bioactive metabolites from macrofungi: Ethnopharmacology, biological activities and chemistry. Fungal Divers. 62:1–40. 2013. View Article : Google Scholar

20 

De Silva DD, Rapior S, Fons F, Bahkali AH and Hyde KD: Medicinal mushrooms in supportive cancer therapies: An approach to anti-cancer effects and putative mechanisms of action. Fungal Divers. 55:1–35. 2012. View Article : Google Scholar

21 

Pao HY, Pan BS, Leu SF and Huang BM: Cordycepin stimulated steroidogenesis in MA-10 mouse Leydig tumor cells through the protein kinase C Pathway. J Agric Food Chem. 60:4905–4913. 2102. View Article : Google Scholar

22 

Kim JR, Yeon SH, Kim HS and Ahn YJ: Larvicidal activity against Plutella xylostella of cordycepin from the fruiting body of Cordyceps militaris. Pest Manag Sci. 58:713–717. 2012. View Article : Google Scholar

23 

Ramesh T, Yoo SK, Kim SW, Hwang SY, Sohn SH, Kim IW and Kim SK: Cordycepin (3′-deoxyadenosine) attenuates age-related oxidative stress and ameliorates antioxidant capacity in rats. Exp Gerontol. 47:979–987. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Guo P, Kai Q, Gao J, Lian ZQ, Wu CM, Wu CA and Zhu HB: Cordycepin prevents hyperlipidemia in hamsters fed a high-fat diet via activation of AMP-activated protein kinase. J Pharm Sci. 113:395–403. 2010. View Article : Google Scholar

25 

Kim SS, Lim KS, Kim HJ, Chong MS, Cho HE, Choi YH, et al: Effects of extracts from mixed culture with Tricholoma Matsutake mycelium and Cordyceps Militaris mycelium on mlood glucose in streptozotocin-induced diabetic rats. Korean J Orient Physiol Pathol. 22:365–370. 2008.

26 

Hung YP and Lee CL: Higher anti-liver fibrosis effect of cordyceps militaris-fermented product cultured with deep ocean water via inhibiting proinflammatory factors and fibrosis-related factors expressions. Mar Drugs. 15(pii): E1682017. View Article : Google Scholar : PubMed/NCBI

27 

Lee MR, Kim JE, Yun WB, Choi JY, Park JJ, Kim HR, Song BR, Choi YW, Kim KM and Hwang DY: Lipolytic effect of novel extracts from mulberry (Morus alba) leaves fermented with Cordyceps militaris in the primary adipocytes derived from SD rats. Lab Anim Res. 33:270–279. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Hofbauer KG, Nicholson JR and Boss O: The obesity epidemic: Current and future pharmacological treatments. Annu Rev Pharmacol Toxicol. 47:565–592. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Sharma B and Henderson DC: Sibutramine: Current status as an anti-obesity drug and its future perspectives. Expert Opin Pharmacother. 9:2161–2173. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Lean ME: Pathophysiology of obesity. Proc Nutr Soc. 59:331–336. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Apostolopoulou M, Savopoulos C, Michalakis K, Coppack S, Dardavessis T and Hatzitolios A: Age, weight and obesity. Maturitas. 71:115–119. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Morimoto C, Kameda K, Tsujita T and Okuda H: Relationships between lipolysis induced by various lipolytic agents and hormone-sensitive lipase in rat fat cells. J Lipid Res. 42:120–127. 2001.PubMed/NCBI

34 

Bray GA: Drug treatment of obesity. Rev Endocr Metab Disord. 2:403–418. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Mayer MA, Höcht C, Puyó A and Taira CA: Recent advances in obesity pharmacotherapy. Curr Clin Pharmacol. 4:53–61. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Vázquez-Vela ME, Torres N and Tovar AR: White adipose tissue as endocrine organ and its role in obesity. Arch Med Res. 39:715–728. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Schoonjans K, Staels B and Auwerx J: The peroxisome proliferator activated receptors (PPARS) and their effects on lipid metabolism and adipocyte differentiation. Biochim Biophys Acta. 1302:93–109. 1996. View Article : Google Scholar : PubMed/NCBI

38 

Rosen ED, Walkey CJ, Puigserver P and Spiegelman BM: Transcriptional regulation of adipogenesis. Genes Dev. 14:1293–1307. 2000.PubMed/NCBI

39 

Kliewer SA and Willson TM: The nuclear receptor PPARgamma-bigger than fat. Curr Opin Genet Dev. 8:576–581. 1998. View Article : Google Scholar : PubMed/NCBI

40 

Spiegelman BM: PPAR-gamma: Adipogenic regulator and thiazolidinedione receptor. Diabetes. 47:507–514. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Boizard M, Le Liepvre X, Lemarchand P, Foufelle F, Ferré P and Dugail I: Obesity-related overexpression of fatty-acid synthase gene in adipose tissue involves sterol regulatory element-binding protein transcription factors. J Biol Chem. 273:29164–29171. 1998. View Article : Google Scholar : PubMed/NCBI

42 

Coe NR and Bernlohr DA: Physiological properties and functions of intracellular fatty acid-binding proteins. Biochim Biophys Acta. 1391:287–306. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Zimmermann R, Strauss JG, Haemmerle G, Schoiswohl G, Birner-Gruenberger R, Riederer M, Lass A, Neuberger G, Eisenhaber F, Hermetter A and Zechner R: Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science. 306:1383–1386. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Haemmerle G, Lass A, Zimmermann R, Gorkiewicz G, Meyer C, Rozman J, Heldmaier G, Maier R, Theussl C, Eder S, et al: Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science. 312:734–737. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Ahmadian M, Duncan RE, Varady KA, Frasson D, Hellerstein MK, Birkenfeld AL, Samuel VT, Shulman GI, Wang Y, Kang C and Sul HS: Adipose overexpression of desnutrin promotes fatty acid use and attenuates diet-induced obesity. Diabetes. 58:855–866. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Tansey JT, Sztalryd C, Hlavin EM, Kimmel AR and Londos C: The central role of perilipin a in lipid metabolism and adipocyte lipolysis. IUBMB Life. 56:379–385. 2004. View Article : Google Scholar : PubMed/NCBI

47 

Marcelin G and Chua S Jr: Contributions of adipocyte lipid metabolism to body fat content and implications for the treatment of obesity. Curr Opin Pharmacol. 10:588–593. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee MR, Kim JE, Choi JY, Park JJ, Kim HR, Song BR, Choi YW, Kim KM, Song H, Hwang DY, Hwang DY, et al: Anti‑obesity effect in high‑fat‑diet‑induced obese C57BL/6 mice: Study of a novel extract from mulberry (Morus alba) leaves fermented with Cordyceps militaris . Exp Ther Med 17: 2185-2193, 2019
APA
Lee, M.R., Kim, J.E., Choi, J.Y., Park, J.J., Kim, H.R., Song, B.R. ... Hwang, D.Y. (2019). Anti‑obesity effect in high‑fat‑diet‑induced obese C57BL/6 mice: Study of a novel extract from mulberry (Morus alba) leaves fermented with Cordyceps militaris . Experimental and Therapeutic Medicine, 17, 2185-2193. https://doi.org/10.3892/etm.2019.7191
MLA
Lee, M. R., Kim, J. E., Choi, J. Y., Park, J. J., Kim, H. R., Song, B. R., Choi, Y. W., Kim, K. M., Song, H., Hwang, D. Y."Anti‑obesity effect in high‑fat‑diet‑induced obese C57BL/6 mice: Study of a novel extract from mulberry (Morus alba) leaves fermented with Cordyceps militaris ". Experimental and Therapeutic Medicine 17.3 (2019): 2185-2193.
Chicago
Lee, M. R., Kim, J. E., Choi, J. Y., Park, J. J., Kim, H. R., Song, B. R., Choi, Y. W., Kim, K. M., Song, H., Hwang, D. Y."Anti‑obesity effect in high‑fat‑diet‑induced obese C57BL/6 mice: Study of a novel extract from mulberry (Morus alba) leaves fermented with Cordyceps militaris ". Experimental and Therapeutic Medicine 17, no. 3 (2019): 2185-2193. https://doi.org/10.3892/etm.2019.7191