1
|
Pahlavani M, Pham K, Kalupahana NS, Morovati A, Ramalingam L, Abidi H, Kiridana V, Moustaid-Moussa N. Thermogenic adipose tissues: Promising therapeutic targets for metabolic diseases. J Nutr Biochem 2025; 137:109832. [PMID: 39653156 DOI: 10.1016/j.jnutbio.2024.109832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
The ongoing increase in the prevalence of obesity and its comorbidities such as cardiovascular disease, type 2 diabetes (T2D) and dyslipidemia warrants discovery of novel therapeutic options for these metabolic diseases. Obesity is characterized by white adipose tissue expansion due to chronic positive energy balance as a result of excessive energy intake and/or reduced energy expenditure. Despite various efforts to prevent or reduce obesity including lifestyle and behavioral interventions, surgical weight reduction approaches and pharmacological methods, there has been limited success in significantly reducing obesity prevalence. Recent research has shown that thermogenic adipocyte (brown and beige) activation or formation, respectively, could potentially act as a therapeutic strategy to ameliorate obesity and its related disorders. This can be achieved through the ability of these thermogenic cells to enhance energy expenditure and regulate circulating levels of glucose and lipids. Thus, unraveling the molecular mechanisms behind the formation and activation of brown and beige adipocytes holds the potential for probable therapeutic paths to combat obesity. In this review, we provide a comprehensive update on the development and regulation of different adipose tissue types. We also emphasize recent interventions in harnessing therapeutic potential of thermogenic adipocytes by bioactive compounds and new pharmacological anti-obesity agents.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Sciences, Texas Woman's University, Dallas, Texas, USA
| | - Kenneth Pham
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Ashti Morovati
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Studies, Syracuse University, Syracuse, New York, USA
| | - Hussain Abidi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Vasana Kiridana
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Institute for One Health Innovation, Texas Tech University and Texas Tech Health Sciences Center, Lubbock, Texas, USA.
| |
Collapse
|
2
|
Kim SH, Park WY, Song G, Park JY, Jung SJ, Ahn KS, Um JY. 4-hydroxybenzoic acid induces browning of white adipose tissue through the AMPK-DRP1 pathway in HFD-induced obese mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156353. [PMID: 39799892 DOI: 10.1016/j.phymed.2024.156353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Beige adipocytes have physiological functions similar to brown adipocytes, which are available to increase energy expenditure through uncoupling protein 1 (UCP1) within mitochondria. Recently, many studies showed white adipocytes can undergo remodeling into beige adipocytes, called "browning", by increasing fusion and fission events referred to as mitochondrial dynamics. PURPOSE In this study, we aimed to investigate the browning effects of 4-hydroxybenzoic acid (4-HA), one of the major compounds of black raspberries. METHODS We examined the mechanism underlying the browning properties of 4-HA focusing on UCP1-dependent non-shivering thermogenesis in 3T3-L1 white adipocytes, high-fat diet (HFD)-induced obese male C57BL/6J mice, and cold-exposed male C57BL/6J mice. RESULTS 4-HA treatment elevates browning markers such as UCP1, T-Box transcription factor 1, and PR domain containing 16, mitochondrial function factors like oxidative phosphorylation complex as well as mitochondrial dynamic-related factors like phosphorylated dynamin-related protein 1 (p-DRP1), DRP1, and mitofusin 1 in 3T3-L1 white adipocytes, which were also confirmed in inguinal white adipose tissue (iWAT) of HFD-induced obese mice. Mdivi-1 blocked the increased DRP1-mediated mitochondrial fission by 4-HA, and even the browning effect of 4-HA was abolished. Furthermore, 4-HA increased AMP-activated protein kinase (AMPK) in both the 3T3-L1 white adipocytes and iWAT of HFD-induced obese mice. Inhibition of AMPK with Compound C also blocked the 4-HA-induced mitochondrial fission and browning effect. CONCLUSIONS 4-HA induces the browning of white adipocytes into beige adipocytes by regulating the DRP1-mediated mitochondrial dynamics through AMPK. These findings suggest that 4-HA could serve as a therapeutic candidate for obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Sang Hee Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 02447, Seoul, South Korea
| | - Woo Yong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Gahee Song
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, 02447, Seoul, South Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Ja Yeon Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 02447, Seoul, South Korea
| | - Se Jin Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 02447, Seoul, South Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 02447, Seoul, South Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 02447, Seoul, South Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, 02447, Seoul, South Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, 02447, Seoul, South Korea.
| |
Collapse
|
3
|
Jee W, Cho HS, Kim SW, Bae H, Chung WS, Cho JH, Kim H, Song MY, Jang HJ. Lycium chinense Mill Induces Anti-Obesity and Anti-Diabetic Effects In Vitro and In Vivo. Int J Mol Sci 2024; 25:8572. [PMID: 39201257 PMCID: PMC11354703 DOI: 10.3390/ijms25168572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
This study investigated the effects of Lycium chinense Mill (LCM) extract on obesity and diabetes, using both in vitro and high-fat diet (HFD)-induced obesity mouse models. We found that LCM notably enhanced glucagon-like peptide-1 (GLP-1) secretion in NCI-h716 cells from 411.4 ± 10.75 pg/mL to 411.4 ± 10.75 pg/mL compared to NT (78.0 ± 0.67 pg/mL) without causing cytotoxicity, implying the involvement of Protein Kinase A C (PKA C) and AMP-activated protein kinase (AMPK) in its action mechanism. LCM also decreased lipid droplets and lowered the expression of adipogenic and lipogenic indicators, such as Fatty Acid Synthase (FAS), Fatty Acid-Binding Protein 4 (FABP4), and Sterol Regulatory Element-Binding Protein 1c (SREBP1c), indicating the suppression of adipocyte differentiation and lipid accumulation. LCM administration to HFD mice resulted in significant weight loss (41.5 ± 3.3 g) compared to the HFD group (45.1 ± 1.8 g). In addition, improved glucose tolerance and serum lipid profiles demonstrated the ability to counteract obesity-related metabolic issues. Additionally, LCM exhibited hepatoprotective properties by reducing hepatic lipid accumulation and diminishing white adipose tissue mass and adipocyte size, thereby demonstrating its effectiveness against hepatic steatosis and adipocyte hypertrophy. These findings show that LCM can be efficiently used as a natural material to treat obesity and diabetes, providing a new approach for remedial and therapeutic purposes.
Collapse
Affiliation(s)
- Wona Jee
- College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (W.J.); (S.W.K.); (H.B.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hong-Seok Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-S.C.); (W.-S.C.); (J.-H.C.); (H.K.)
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seok Woo Kim
- College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (W.J.); (S.W.K.); (H.B.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hanbit Bae
- College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (W.J.); (S.W.K.); (H.B.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Won-Seok Chung
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-S.C.); (W.-S.C.); (J.-H.C.); (H.K.)
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae-Heung Cho
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-S.C.); (W.-S.C.); (J.-H.C.); (H.K.)
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyungsuk Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-S.C.); (W.-S.C.); (J.-H.C.); (H.K.)
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mi-Yeon Song
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-S.C.); (W.-S.C.); (J.-H.C.); (H.K.)
- Department of Korean Rehabilitation Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea; (W.J.); (S.W.K.); (H.B.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
4
|
Bellitto V, Gabrielli MG, Martinelli I, Roy P, Nittari G, Cocci P, Palermo FA, Amenta F, Micioni Di Bonaventura MV, Cifani C, Tomassoni D, Tayebati SK. Dysfunction of the Brown Adipose Organ in HFD-Obese Rats and Effect of Tart Cherry Supplementation. Antioxidants (Basel) 2024; 13:388. [PMID: 38671836 PMCID: PMC11047636 DOI: 10.3390/antiox13040388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity has a great impact on adipose tissue biology, based on its function as a master regulator of energy balance. Brown adipose tissue (BAT) undergoes remodeling, and its activity declines in obese subjects due to a whitening process. The anti-obesity properties of fruit extracts have been reported. The effects of tart cherry against oxidative stress, inflammation, and the whitening process in the BAT of obese rats were investigated. Intrascapular BAT (iBAT) alterations and effects of Prunus cerasus L. were debated in rats fed for 17 weeks with a high-fat diet (DIO), in DIO supplemented with seed powder (DS), and with seed powder plus the juice (DJS) of tart cherry compared to CHOW rats fed with a normo-caloric diet. iBAT histologic observations revealed a whitening process in DIO rats that was reduced in the DS and DJS groups. A modulation of uncoupling protein-1 (UCP-1) protein and gene expression specifically were detected in the obese phenotype. An upregulation of UCP-1 and related thermogenic genes after tart cherry intake was detected compared to the DIO group. Metabolic adjustment, endoplasmic reticulum stress, protein carbonylation, and the inflammatory microenvironment in the iBAT were reported in DIO rats. The analysis demonstrated an iBAT modulation that tart cherry promoted. In addition to our previous results, these data confirm the protective impact of tart cherry consumption on obesity.
Collapse
Affiliation(s)
- Vincenzo Bellitto
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Maria Gabriella Gabrielli
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Ilenia Martinelli
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Proshanta Roy
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Giulio Nittari
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Paolo Cocci
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Francesco Alessandro Palermo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Francesco Amenta
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Maria Vittoria Micioni Di Bonaventura
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Carlo Cifani
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (M.G.G.); (P.C.); (F.A.P.); (D.T.)
| | - Seyed Khosrow Tayebati
- School of Medicinal Sciences and Health Products, University of Camerino, 62032 Camerino, Italy; (V.B.); (I.M.); (P.R.); (G.N.); (F.A.); (M.V.M.D.B.); (C.C.)
| |
Collapse
|
5
|
Liu X, Zhang Z, Song Y, Xie H, Dong M. An update on brown adipose tissue and obesity intervention: Function, regulation and therapeutic implications. Front Endocrinol (Lausanne) 2022; 13:1065263. [PMID: 36714578 PMCID: PMC9874101 DOI: 10.3389/fendo.2022.1065263] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
Overweight and obesity have become a world-wide problem. However, effective intervention approaches are limited. Brown adipose tissue, which helps maintain body temperature and contributes to thermogenesis, is dependent on uncoupling protein1. Over the last decade, an in-creasing number of studies have found that activating brown adipose tissue and browning of white adipose tissue can protect against obesity and obesity-related metabolic disease. Brown adipose tissue has gradually become an appealing therapeutic target for the prevention and re-versal of obesity. However, some important issues remain unresolved. It is not certain whether increasing brown adipose tissue activity is the cause or effect of body weight loss or what the risks might be for sympathetic nervous system-dependent non-shivering thermogenesis. In this review, we comprehensively summarize approaches to activating brown adipose tissue and/or browning white adipose tissue, such as cold exposure, exercise, and small-molecule treatment. We highlight the functional mechanisms of small-molecule treatment and brown adipose tissue transplantation using batokine, sympathetic nervous system and/or gut microbiome. Finally, we discuss the causality between body weight loss induced by bariatric surgery, exercise, and brown adipose tissue activity.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhi Zhang
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yajie Song
- Institute of Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, Henan, China
| | - Hengchang Xie
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- *Correspondence: Meng Dong, ; Hengchang Xie,
| | - Meng Dong
- Department of Nutrition and Food Hygiene, College of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Meng Dong, ; Hengchang Xie,
| |
Collapse
|
6
|
Lee MK, Lee B, Kim CY. Natural Extracts That Stimulate Adipocyte Browning and Their Underlying Mechanisms. Antioxidants (Basel) 2021; 10:antiox10020308. [PMID: 33671335 PMCID: PMC7922619 DOI: 10.3390/antiox10020308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/05/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022] Open
Abstract
Despite progress in understanding the developmental lineage and transcriptional factors regulating brown and beige adipocytes, the role of environmental modifiers, such as food components and natural extracts, remains to be elucidated. Furthermore, the undesirable pleiotropic effects produced by synthetic drugs targeting adipose tissue browning and thermogenesis necessitate research into alternative natural sources to combat obesity and related metabolic disorders. The current review, therefore, focused on the effects of various extracts from foods, plants, and marine products on adipose tissue browning and obesity. In particular, the recent findings of food components and marine products on adipose tissue browning will be discussed here.
Collapse
Affiliation(s)
- Min-Kyeong Lee
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Daeyeon Dong, Busan 608737, Korea;
| | - Bonggi Lee
- Department of Food Science and Nutrition, Pukyong National University, Nam-gu, Daeyeon Dong, Busan 608737, Korea;
- Correspondence: (B.L.); (C.Y.K.); Tel.: +82-51-629-5852 (B.L.); +82-53-810-2871 (C.Y.K.)
| | - Choon Young Kim
- Department of Food and Nutrition, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea
- Correspondence: (B.L.); (C.Y.K.); Tel.: +82-51-629-5852 (B.L.); +82-53-810-2871 (C.Y.K.)
| |
Collapse
|
7
|
Cho SY, Lim S, Ahn KS, Kwak HJ, Park J, Um JY. Farnesol induces mitochondrial/peroxisomal biogenesis and thermogenesis by enhancing the AMPK signaling pathway in vivo and in vitro. Pharmacol Res 2021; 163:105312. [PMID: 33246168 DOI: 10.1016/j.phrs.2020.105312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/30/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022]
Abstract
Thermogenic activation of brown adipose tissue has been considered as an obesity treatment strategy that consumes energy. In this study, we investigated whether farnesol in vivoandin vitro models induces thermogenesis and affect the activation of the mitochondria and peroxisomes, which are key organelles in activated brown adipocytes. Farnesol induced the expression of thermogenic factors such as uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor γ coactivator 1 alpha (PGC1α), and PR domain zinc-finger protein 16 (PRDM16) together with the phosphorylation of AMP-activated protein kinase alpha (AMPKα) in brown adipose tissue and primary cultured brown adipocytes. Farnesol promoted lipolytic enzymes: hormone sensitive lipase (HSL) and adipose triglyceride lipase (ATGL). We confirmed that these inductions of lipolysis by farnesol were the underlying causes of β-oxidation activation. Farnesol also increased the expression of oxidative phosphorylation (OXPHOS) complexes and the oxygen consumption rate (OCR) and the expansion of peroxisomes. Moreover, we proved that the thermogenic activity of farnesol was dependent on AMPKα activation using Compound C inhibitor or siRNA-AMPKα knockdown. These results suggest that farnesol may be a potential agent for the treatment of obesity by inducing energy consumption through heat generation.
Collapse
Affiliation(s)
- Seon Yeon Cho
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Basic Research Laboratory for Comorbidity Research and Department of Comorbidity Research, KyungHee Institute of Convergence Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seona Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Basic Research Laboratory for Comorbidity Research and Department of Comorbidity Research, KyungHee Institute of Convergence Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kwang Seok Ahn
- Basic Research Laboratory for Comorbidity Research and Department of Comorbidity Research, KyungHee Institute of Convergence Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Life Science, College of Natural Sciences, Kyonggi University, Suwon, Republic of Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Basic Research Laboratory for Comorbidity Research and Department of Comorbidity Research, KyungHee Institute of Convergence Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
8
|
A phytoestrogen secoisolariciresinol diglucoside induces browning of white adipose tissue and activates non-shivering thermogenesis through AMPK pathway. Pharmacol Res 2020; 158:104852. [PMID: 32438038 DOI: 10.1016/j.phrs.2020.104852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 01/07/2023]
Abstract
Secoisolariciresinol diglucoside (SDG) is the main phytoestrogen component of flaxseed known as an antioxidant. Current study focused on the effect of SDG in white adipose tissue (WAT) browning. Browning of WAT is considered as a promising treatment strategy for metabolic diseases. To demonstrate the effect of SDG as an inducer of browning, brown adipocyte markers were investigated in inguinal WAT (iWAT) of high fat diet-fed obese mice and genetically obese db/db mice after SDG administration. SDG increased thermogenic factors such as uncoupling protein 1, peroxisome proliferator-activated receptor gamma coactivator 1 alpha and PR domain containing 16 in iWAT and brown adipose tissue (BAT) of mice. Similar results were shown in beige-induced 3T3-L1 adipocytes and primary cultured brown adipocytes. Furthermore, SDG increased factors of mitochondrial biogenesis and activation. We also observed SDG-induced alteration of AMP-activated protein kinase α (AMPKα). As AMPKα is closely related in the regulation of adipogenesis and thermogenesis, we then evaluated the effect of SDG in AMPKα-inhibited conditions. Genetic or chemical inhibition of AMPKα demonstrated that the role of SDG on browning and thermogenesis was dependent on AMPKα signaling. In conclusion, our data suggest SDG as a potential candidate for improvement of obesity and other metabolic disorders.
Collapse
|
9
|
Zu YX, Lu HY, Liu WW, Jiang XW, Huang Y, Li X, Zhao QC, Xu ZH. Jiang Gui Fang activated interscapular brown adipose tissue and induced epididymal white adipose tissue browning through the PPARγ/SIRT1-PGC1α pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112271. [PMID: 31586693 DOI: 10.1016/j.jep.2019.112271] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/28/2019] [Accepted: 10/02/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gui Zhi Tang, a well-known Chinese herbal formula recorded in the Eastern Han Dynasty, has been widely used to treat exogenous cold for thousands of years. Recent studies have shown that Gui Zhi Tang has the effect of regulating the body temperature. Because of its effect on heat production, protecting vital organs of the body and avoiding damage from the cold environment, Jiang Gui Fang (JG) was obtained from the Department of Traditional Chinese Medicine at the General Hospital of Northern Theatre Command where it has been used clinically for many years and has exhibited favourable efficacy. Based on research on Gui Zhi Tang, the principles of traditional Chinese medicine and survey of a large number of studies, this empirical formula was developed. The composition of JG included Dried ginger, Cassia twig, and Liquorice in Gui Zhi Tang, which play a major role in the treatment of exogenous cold, and combined these components with other Chinese medicines, such as Pueraria, Spatholobus, Acanthopanacis cortex, Evodiae fructus, and Codonopsis pilosula. AIM OF THE STUDY To promote the core body temperature and prevent invasion of the major organs from the cold environment, we studied the effect of JG on the core body temperature of mice and then explored its regulation of interscapular brown adipose tissue (iBAT) and epididymal white adipose tissue (eWAT) and the possible mechanism. Finally, we determined the phytochemical composition of JG that plays a role in heat production. MATERIALS AND METHODS In vivo study, we performed a 4-week treatment of JG in acute cold environment at -20 °C and chronic cold exposure at 4 °C. The core temperature, adipose tissue weight, serum parameters, and morphological observation of adipocytes, liver and kidney were measured. Then we investigated the expression levels of adipogenic factors, thermogenic factors and lipoprotein. In vitro, we determined the lipid droplet content, ATP content, and the maximum oxygen consumption of mitochondria. RESULTS JG treatment promoted core temperature, inhibited eWAT weight, protected liver, and reduced glucose and lipids in Kunming (KM) mice. JG also increased the expression of BAT-associated thermogenic factors, including uncoupling protein 1 (UCP1) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC1α). The levels of the lipogenic factor peroxisome proliferate-activator receptor gamma (PPARγ) and the lipolytic protein hormone-sensitive triglyceride lipase (HSL) in eWAT were elevated. The results of H&E and immunohistochemistry showed that JG significantly reduced the size of iBAT and eWAT and increased the content of UCP1. In vitro, JG reduced the content of lipid droplets and ATP in brown fat cells. The maximum oxygen consumption capacity of mitochondria and the expression levels of UCP1, PGC1α and silent mating type information regulation 2 homologue 1 (SIRT1) were enhanced after JG treatment. CONCLUSIONS In vivo and in vitro studies, the results demonstrated that JG obviously increased the core temperature of mice by activating iBAT and inducing eWAT browning, which proved the mechanism is closely related to the PPARγ/SIRT1- PGC1α pathway. In this paper, we will provide a reference for further study of iBAT activation and eWAT browning.
Collapse
Affiliation(s)
- Yu-Xin Zu
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Hong-Yuan Lu
- School of Life Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Wen-Wu Liu
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Xiao-Wen Jiang
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Yuan Huang
- School of Life Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Xiang Li
- School of Life Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Qing-Chun Zhao
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China.
| | - Zi-Hua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
10
|
Wang N, Lu HY, Li X, Du YJ, Meng WH, Ding HW, Zhao QC. ZW290 Increases Cold Tolerance by Inducing Thermogenesis via the Upregulation of Uncoupling Protein 1 in Brown Adipose Tissue In Vitro and In Vivo. Lipids 2020; 54:265-276. [PMID: 31087415 DOI: 10.1002/lipd.12148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 03/10/2019] [Accepted: 03/26/2019] [Indexed: 11/06/2022]
Abstract
To provide molecular evidence on the thermogenic mechanism of primary brown adipocytes, western blot analysis was used to detect brown adipose tissue (BAT)-specific gene expressions. BAT protects the mammals from hypothermia injury with a large amount of mitochondria and high expression of uncoupling Protein 1 (UCP1), which is the vital protein to determine the heat production in BAT. In our previous study, the compound ZW290 (the structure shown in Fig. 1) was obtained by molecular docking with a UCP1 inducer. In the present study, ZW290 not only significantly upregulated the expression of UCP1 protein (p < 0.01) and its related signaling pathway in the primary brown adipocytes, but also remarkably decreased the mitochondrial membrane potential and the concentration of adenosine triphosphate (ATP) (p < 0.01). Kunming (KM) mice were kept under acute cold exposure (-20°C) to evaluate the preventive and protective effects of ZW290 on cold injury, and revealed its regulating mechanism in vitro. The rectal and body temperatures of ZW290-treated mice were significantly higher than those of the control (or model) group both at room temperature and at -20°C (p < 0.001). Hematoxylin-eosin (HE) staining and immunohistochemistry indicated that ZW290 notably decreased the size of lipid droplets in BAT and increased the content of mitochondria and the expression of UCP1 in BAT and white adipose tissue (WAT). Furthermore, the survival rate showed that ZW290 could prolong the overall survival of mice. Therefore, we obtained the conclusion that ZW290 might transform energy into heat by inhibiting ATP synthesis and increasing the expression of UCP1. Additionally, ZW290 may enhance cold tolerance by increasing heat production through increasing the content of mitochondria and the expression of UCP1 in BAT and WAT.
Collapse
Affiliation(s)
- Nan Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Hong-Yuan Lu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China
| | - Ya-Jie Du
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Wei-Hong Meng
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China
| | - Huai-Wei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| | - Qing-Chun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Wenhua Road No. 83, Shenhe District, Shenyang 110840, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, Wenhua Road No. 103, Shenhe District, Shenyang 110016, China
| |
Collapse
|
11
|
Black Raspberry ( Rubus coreanus Miquel) Promotes Browning of Preadipocytes and Inguinal White Adipose Tissue in Cold-Induced Mice. Nutrients 2019; 11:nu11092164. [PMID: 31509935 PMCID: PMC6769844 DOI: 10.3390/nu11092164] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
The alteration of white adipose tissue (WAT) "browning", a change of white into beige fat, has been considered as a new therapeutic strategy to treat obesity. In this study, we investigated the browning effect of black raspberry (Rubus coreanus Miquel) using in vitro and in vivo models. Black raspberry water extract (BRWE) treatment inhibited lipid accumulation in human mesenchymal stem cells (hMSCs) and zebrafish. To evaluate the thermogenic activity, BRWE was orally administered for 2 weeks, and then, the mice were placed in a 4 °C environment. As a result, BRWE treatment increased rectal temperature and inguinal WAT (iWAT) thermogenesis by inducing the expression of beige fat specific markers such as PR domain zinc-finger protein 16 (PRDM16), uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), and t-box protein 1 (TBX1) in cold-exposed mice. Furthermore, ellagic acid (EA), a constituent of BRWE, markedly promoted beige specific markers: UCP1, PGC1α, TBX1, and nuclear respiratory factor 1 in beige differentiation media (DM)-induced 3T3-L1 adipocytes. Our findings indicate that BRWE can promote beige differentiation/activation, and EA is the active compound responsible for such effect. Thus, we suggest the nature-derived agents BRWE and EA as potential agents for obesity treatment.
Collapse
|
12
|
Synergistic Effect of Bupleuri Radix and Scutellariae Radix on Adipogenesis and AMP-Activated Protein Kinase: A Network Pharmacological Approach. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5269731. [PMID: 30210572 PMCID: PMC6126083 DOI: 10.1155/2018/5269731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/11/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022]
Abstract
Obesity has become a major health threat in developed countries. However, current medications for obesity are limited because of their adverse effects. Interest in natural products for the treatment of obesity is thus rapidly growing. Korean medicine is characterized by the wide use of herbal formulas. However, the combination rule of herbal formulas in Korean medicine lacks experimental evidence. According to Shennong's Classic of Materia Medica, the earliest book of herbal medicine, Bupleuri Radix (BR) and Scutellariae Radix (SR) possess the Sangsoo relationship, which means they have synergistic features when used together. Therefore these two are frequently used together in prescriptions such as Sosiho-Tang. In this study, we used the network pharmacological method to predict the interaction between these two herbs and then investigated the effects of BR, SR, and their combination on obesity in 3T3-L1 adipocytes. BR, SR, and BR-SR mixture significantly decreased lipid accumulation and the expressions of two major adipogenic factors, peroxisome proliferator-activated receptor-gamma (PPARγ) and CCAAT/enhancer-binding protein-alpha (C/EBPα), and their downstream genes, Adipoq, aP2, and Lipin1 in 3T3-L1 cells. In addition, the BR-SR mixture had synergistic effects compared with BR or SR on inhibition of adipogenic-gene expressions. BR and SR also inhibited the protein expressions of PPARγ and C/EBPα. Furthermore, the two extracts successfully activated AMP-activated protein kinase alpha (AMPK α), the key regulator of energy metabolism. When compared to those of BR or SR, the BR-SR mixture showed higher inhibition rates of PPARγ and C/EBPα, along with higher activation rate of AMPK. These results indicate a new potential antiobese pharmacotherapy and also provide scientific evidence supporting the usage of herbal combinations instead of mixtures in Korean medicine.
Collapse
|
13
|
Jung Y, Park J, Kim HL, Sim JE, Youn DH, Kang J, Lim S, Jeong MY, Yang WM, Lee SG, Ahn KS, Um JY. Vanillic acid attenuates obesity via activation of the AMPK pathway and thermogenic factors in vivo and in vitro. FASEB J 2018; 32:1388-1402. [PMID: 29141998 DOI: 10.1096/fj.201700231rr] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Energy expenditure is a target gaining recent interest for obesity treatment. The antiobesity effect of vanillic acid (VA), a well-known flavoring agent, was investigated in vivo and in vitro. High-fat diet (HFD)-induced obese mice and genetically obese db/db mice showed significantly decreased body weights after VA administration. Two major adipogenic markers, peroxisome proliferator activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα), were reduced while the key factor of energy metabolism, AMPKα, was increased in the white adipose tissue and liver tissue of VA-treated mice. Furthermore, VA inhibited lipid accumulation and reduced hepatotoxic/inflammatory markers in liver tissues of mice and HepG2 hepatocytes. VA treatment also decreased differentiation of 3T3-L1 adipocytes by regulating adipogenic factors including PPARγ and C/EBPα. AMPKα small interfering RNA was used to examine whether AMPK was associated with the actions of VA. In AMPKα-nulled 3T3-L1 cells, the inhibitory action of VA on PPARγ and C/EBPα was attenuated. Furthermore, in brown adipose tissues of mice and primary cultured brown adipocytes, VA increased mitochondria- and thermogenesis-related factors such as uncoupling protein 1 and PPARγ-coactivator 1-α. Taken together, our results suggest that VA has potential as an AMPKα- and thermogenesis-activating antiobesity agent.-Jung, Y., Park, J., Kim, H.-L., Sim, J.-E., Youn, D.-H., Kang, J., Lim, S., Jeong, M.-Y., Yang, W. M., Lee, S.-G., Ahn, K. S., Um, J.-Y. Vanillic acid attenuates obesity via activation of the AMPK pathway and thermogenic factors in vivo and in vitro.
Collapse
Affiliation(s)
- Yunu Jung
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Jinbong Park
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Hye-Lin Kim
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Jung-Eun Sim
- Department of Biological Sciences in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Dong-Hyun Youn
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - JongWook Kang
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Seona Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Mi-Young Jeong
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Woong Mo Yang
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Seok-Geun Lee
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Kwang Seok Ahn
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| | - Jae-Young Um
- College of Korean Medicine and Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
14
|
Secoisolariciresinol diglucoside inhibits adipogenesis through the AMPK pathway. Eur J Pharmacol 2018; 820:235-244. [DOI: 10.1016/j.ejphar.2017.12.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/15/2017] [Accepted: 12/15/2017] [Indexed: 11/18/2022]
|
15
|
Kim HL, Jung Y, Park J, Youn DH, Kang J, Lim S, Lee BS, Jeong MY, Choe SK, Park R, Ahn KS, Um JY. Farnesol Has an Anti-obesity Effect in High-Fat Diet-Induced Obese Mice and Induces the Development of Beige Adipocytes in Human Adipose Tissue Derived-Mesenchymal Stem Cells. Front Pharmacol 2017; 8:654. [PMID: 29033835 PMCID: PMC5627035 DOI: 10.3389/fphar.2017.00654] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/01/2017] [Indexed: 12/17/2022] Open
Abstract
Brown adipocytes dissipate energy as heat and hence have an important therapeutic capacity for obesity. Development of brown-like adipocytes (also called beige) is also another attractive target for obesity treatment. Here, we investigated the effect of farnesol, an isoprenoid, on adipogenesis in adipocytes and on the browning of white adipose tissue (WAT) as well as on the weight gain of high-fat diet (HFD)-induced obese mice. Farnesol inhibited adipogenesis and the related key regulators including peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α through the up-regulation of AMP-activated protein kinase in 3T3-L1 murine adipocytes and human adipose tissue-derived mesenchymal stem cells (hAMSCs). Farnesol markedly increased the expression of uncoupling protein 1 and PPARγ coactivator 1 α in differentiated hAMSCs. In addition, farnesol limited the weight gain in HFD obese mice and induced the development of beige adipocytes in both inguinal and epididymal WAT. These results suggest that farnesol could be a potential therapeutic agent for obesity treatment.
Collapse
Affiliation(s)
- Hye-Lin Kim
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Yunu Jung
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Jinbong Park
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Dong-Hyun Youn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - JongWook Kang
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Seona Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Beom Su Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Mi-Young Jeong
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Seong-Kyu Choe
- Department of Microbiology and Center for Metabolic Function Regulation, School of Medicine, Wonkwang UniversityIksan, South Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Jae-Young Um
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| |
Collapse
|
16
|
Lee MY, Kim HY, Lee DE, Singh D, Yeo SH, Baek SY, Park YK, Lee CH. Construing temporal metabolomes for acetous fermentative production of Rubus coreanus vinegar and its in vivo nutraceutical effects. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.04.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
17
|
Rao Y, Yu H, Gao L, Lu YT, Xu Z, Liu H, Gu LQ, Ye JM, Huang ZS. Natural alkaloid bouchardatine ameliorates metabolic disorders in high-fat diet-fed mice by stimulating the sirtuin 1/liver kinase B-1/AMPK axis. Br J Pharmacol 2017; 174:2457-2470. [PMID: 28493443 DOI: 10.1111/bph.13855] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/09/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Promoting energy metabolism is known to provide therapeutic effects for obesity and associated metabolic disorders. The present study evaluated the therapeutic effects of the newly identified bouchardatine (Bou) on obesity-associated metabolic disorders and the molecular mechanisms of these effects. EXPERIMENTAL APPROACH The molecular mode of action of Bou for its effects on lipid metabolism was first examined in 3T3-L1 adipocytes and HepG2 cells. This was followed by an evaluation of its metabolic effects in mice fed a high-fat diet for 16 weeks with Bou being administered in the last 5 weeks. Further mechanistic investigations were conducted in pertinent organs of the mice and relevant cell models. KEY RESULTS In 3T3-L1 adipocytes, Bou reduced lipid content and increased sirtuin 1 (SIRT1) activity to facilitate liver kinase B1 (LKB1) activation of AMPK. Chronic administration of Bou (50 mg∙kg-1 every other day) in mice significantly attenuated high-fat diet-induced increases in body weight gain, dyslipidaemia and fatty liver without affecting food intake and no adverse effects were detected. These metabolic effects were associated with activation of the SIRT1-LKB1-AMPK signalling pathway in adipose tissue and liver. Of particular note, UCP1 expression and mitochondrial biogenesis were increased in both white and brown adipose tissues of Bou-treated mice. Incubation with Bou induced similar changes in primary brown adipocytes isolated from mice. CONCLUSIONS AND IMPLICATIONS Bou may have therapeutic potential for obesity-related metabolic diseases by increasing the capacity of energy expenditure in adipose tissues and liver through a mechanism involving the SIRT1-LKB1-AMPK axis.
Collapse
Affiliation(s)
- Yong Rao
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hong Yu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lin Gao
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yu-Ting Lu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhao Xu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hong Liu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lian-Quan Gu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ji-Ming Ye
- Molecular Pharmacology for Diabetes Group, School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Zhi-Shu Huang
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Su HM, Feng LN, Zheng XD, Chen W. Myricetin protects against diet-induced obesity and ameliorates oxidative stress in C57BL/6 mice. J Zhejiang Univ Sci B 2017; 17:437-46. [PMID: 27256677 DOI: 10.1631/jzus.b1600074] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Myricetin is a naturally occurring antioxidant commonly found in various plants. However, little information is available with respect to its direct anti-obesity effects. OBJECTIVE This study was undertaken to investigate the effect of myricetin on high-fat diet (HFD)-induced obesity in C57BL/6 mice. RESULTS Administration of myricetin dramatically reduced the body weight of diet-induced obese mice compared with solely HFD-induced mice. Several parameters related to obesity including serum glucose, triglyceride, and cholesterol were significantly decreased in myricetin-treated mice. Moreover, obesity-associated oxidative stress (glutathione peroxidase (GPX) activity, total antioxidant capacity (T-AOC), and malondialdehyde (MDA)) and inflammation (tumor necrosis factor-α (TNF-α)) were ameliorated in myricetin-treated mice. Further investigation revealed that the protective effect of myricetin against HFD-induced obesity in mice appeared to be partially mediated through the down-regulation of mRNA expression of adipogenic transcription factors peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα), and lipogenic transcription factor sterol regulatory element-binding protein 1c (SREBP-1c). CONCLUSIONS Consumption of myricetin may help to prevent obesity and obesity-related metabolic complications.
Collapse
Affiliation(s)
- Hong-Ming Su
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Li-Na Feng
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, China
| | - Xiao-Dong Zheng
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Wei Chen
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
19
|
Bonet ML, Mercader J, Palou A. A nutritional perspective on UCP1-dependent thermogenesis. Biochimie 2017; 134:99-117. [DOI: 10.1016/j.biochi.2016.12.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/23/2016] [Indexed: 12/16/2022]
|
20
|
Lee Y, Kim J, An J, Lee S, Lee H, Kong H, Song Y, Choi HR, Kwon JW, Shin D, Lee CK, Kim K. Restoration of Declined Immune Responses and Hyperlipidemia by Rubus occidenalis in Diet-Induced Obese Mice. Biomol Ther (Seoul) 2017; 25:140-148. [PMID: 27737523 PMCID: PMC5340538 DOI: 10.4062/biomolther.2016.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/17/2016] [Accepted: 06/14/2016] [Indexed: 01/21/2023] Open
Abstract
Hyperlipidemia, which is closely associated with a fatty diet and aging, is commonly observed in the western and aged society. Therefore, a novel therapeutic approach for this disease is critical, and an immunological view has been suggested as a novel strategy, because hyperlipidemia is closely associated with inflammation and immune dysfunction. In this study, the effects of an aqueous extract of Rubus occidentalis (RO) in obese mice were investigated using immunological indexes. The mice were fed a high-fat diet (HFD) to induce hyperlipidemia, which was confirmed by biochemical analysis and examination of the mouse physiology. Two different doses of RO and rosuvastatin, a cholesterol synthesis inhibitor used as a control, were orally administered. Disturbances in immune cellularity as well as lymphocyte proliferation and cytokine production were significantly normalized by oral administration of RO, which also decreased the elevated serum tumor necrosis factor (TNF)-α level and total cholesterol. The specific immune-related actions of RO comprised considerable improvement in cytotoxic T cell killing functions and regulation of antibody production to within the normal range. The immunological evidence confirms the significant cholesterol-lowering effect of RO, suggesting its potential as a novel therapeutic agent for hyperlipidemia and associated immune decline.
Collapse
Affiliation(s)
- Youngjoo Lee
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Jiyeon Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Jinho An
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Sungwon Lee
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Heetae Lee
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Hyunseok Kong
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Hye Ran Choi
- Berry and Biofood Research Institute, Jeonbuk 56417, Republic of Korea
| | - Ji-Wung Kwon
- Berry and Biofood Research Institute, Jeonbuk 56417, Republic of Korea
| | - Daekeun Shin
- Berry and Biofood Research Institute, Jeonbuk 56417, Republic of Korea
| | - Chong-Kil Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Kyungjae Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
21
|
Lim H, Park J, Kim HL, Kang J, Jeong MY, Youn DH, Jung Y, Kim YI, Kim HJ, Ahn KS, Kim SJ, Choe SK, Hong SH, Um JY. Chrysophanic Acid Suppresses Adipogenesis and Induces Thermogenesis by Activating AMP-Activated Protein Kinase Alpha In vivo and In vitro. Front Pharmacol 2016; 7:476. [PMID: 28008317 PMCID: PMC5143616 DOI: 10.3389/fphar.2016.00476] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022] Open
Abstract
Chrysophanic acid (CA) is a member of the anthraquinone family abundant in rhubarb, a widely used herb for obesity treatment in Traditional Korean Medicine. Though several studies have indicated numerous features of CA, no study has yet reported the effect of CA on obesity. In this study, we tried to identify the anti-obesity effects of CA. By using 3T3-L1 adipocytes and primary cultured brown adipocytes as in vitro models, high-fat diet (HFD)-induced obese mice, and zebrafish as in vivo models, we determined the anti-obesity effects of CA. CA reduced weight gain in HFD-induced obese mice. They also decreased lipid accumulation and the expressions of adipogenesis factors including peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (C/EBPα) in 3T3-L1 adipocytes. In addition, uncoupling protein 1 (UCP1) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), the brown fat specific thermogenic genes, were up-regulated in brown adipocytes by CA treatment. Furthermore, when co-treated with Compound C, the AMP-activated protein kinase (AMPK) inhibitor, the action of CA on AMPKα was nullified in both types of adipocytes, indicating the multi-controlling effect of CA was partially via the AMPKα pathway. Given all together, these results indicate that CA can ameliorate obesity by controlling the adipogenic and thermogenic pathway at the same time. On these bases, we suggest the new potential of CA as an anti-obese pharmacotherapy.
Collapse
Affiliation(s)
- Hara Lim
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Jinbong Park
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Hye-Lin Kim
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - JongWook Kang
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Mi-Young Jeong
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Dong-Hyun Youn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Yunu Jung
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Yong-Il Kim
- Department of Microbiology and Center for Metabolic Function Regulation, School of Medicine, Wonkwang University Iksan, South Korea
| | - Hyun-Ju Kim
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| | - Su-Jin Kim
- Department of Cosmeceutical Science, Daegu Haany University Kyungsan, South Korea
| | - Seong-Kyu Choe
- Department of Microbiology and Center for Metabolic Function Regulation, School of Medicine, Wonkwang University Iksan, South Korea
| | - Seung-Heon Hong
- Department of Pharmacology, College of Pharmacy, Wonkwang University Iksan, South Korea
| | - Jae-Young Um
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation Kyung Hee University, Seoul, South Korea
| |
Collapse
|
22
|
Choi HM, Jung Y, Park J, Kim HL, Youn DH, Kang J, Jeong MY, Lee JH, Yang WM, Lee SG, Ahn KS, Um JY. Cinnamomi Cortex (Cinnamomum verum) Suppresses Testosterone-induced Benign Prostatic Hyperplasia by Regulating 5α-reductase. Sci Rep 2016; 6:31906. [PMID: 27549514 PMCID: PMC4994048 DOI: 10.1038/srep31906] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/26/2016] [Indexed: 01/13/2023] Open
Abstract
Cinnamomi cortex (dried bark of Cinnamomum verum) is an important drug in Traditional Korean Medicine used to improve blood circulation and Yang Qi. Benign prostatic hyperplasia (BPH) is a common chronic disease in aging men. This study was conducted to determine the effect of Cinnamomi cortex water extract (CC) on BPH. BPH was induced by a pre-4-week daily injection of testosterone propionate (TP). Six weeks of further injection with (a) vehicle, (b) TP, (c) TP + CC, (d) TP + finasteride (Fi) was carried on. As a result, the prostate weight and prostatic index of the CC treatment group were reduced. Histological changes including epithelial thickness and lumen area were recovered as normal by CC treatment. The protein expressions of prostate specific antigen, estrogen receptor α (ERα), androgen receptor (AR), 5α-reductase (5AR), and steroid receptor coactivator 1 were suppressed by treatment of CC. Immunohistochemical assays supported the western blot results, as the expressions of AR and ERα were down-regulated by CC treatment as well. Further in vitro experiments showed CC was able to inhibit proliferation of RWPE-1 cells by suppressing 5AR and AR. These results all together suggest CC as a potential treatment for BPH.
Collapse
Affiliation(s)
- Hyun-Myung Choi
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Yunu Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Jinbong Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Hye-Lin Kim
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Dong-Hyun Youn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - JongWook Kang
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Mi-Young Jeong
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Jong-Hyun Lee
- College of Pharmacy, Dongduk Women's University, 60 Hwarang-ro 13-gil, Seongbuk-gu, Seoul, 136-714, Republic of Korea
| | - Woong Mo Yang
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Seok-Geun Lee
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| | - Jae-Young Um
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-Gu, Seoul, 130-701, Republic of Korea
| |
Collapse
|
23
|
Cell Models and Their Application for Studying Adipogenic Differentiation in Relation to Obesity: A Review. Int J Mol Sci 2016; 17:ijms17071040. [PMID: 27376273 PMCID: PMC4964416 DOI: 10.3390/ijms17071040] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 02/08/2023] Open
Abstract
Over the last several years, the increasing prevalence of obesity has favored an intense study of adipose tissue biology and the precise mechanisms involved in adipocyte differentiation and adipogenesis. Adipocyte commitment and differentiation are complex processes, which can be investigated thanks to the development of diverse in vitro cell models and molecular biology techniques that allow for a better understanding of adipogenesis and adipocyte dysfunction associated with obesity. The aim of the present work was to update the different animal and human cell culture models available for studying the in vitro adipogenic differentiation process related to obesity and its co-morbidities. The main characteristics, new protocols, and applications of the cell models used to study the adipogenesis in the last five years have been extensively revised. Moreover, we depict co-cultures and three-dimensional cultures, given their utility to understand the connections between adipocytes and their surrounding cells in adipose tissue.
Collapse
|
24
|
Han YH, Kee JY, Park J, Kim DS, Shin S, Youn DH, Kang J, Jung Y, Lee YM, Park JH, Kim SJ, Um JY, Hong SH. Lipin1-Mediated Repression of Adipogenesis by Rutin. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:565-78. [PMID: 27109161 DOI: 10.1142/s0192415x16500312] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Rutin, also called rutoside or quercetin-3-O-rutinoside and sophorin, is a glycoside between the flavonol quercetin and the disaccharide rutinose. Although many effects of rutin have been reported in vitro and in vivo, the anti-adipogenic effects of rutin have not been fully reported. The aim of this study was to confirm how rutin regulates adipocyte related factors. In this study, rutin decreased the expressions of adipogenesis-related genes, including peroxisome proliferators, activated receptor [Formula: see text] (PPAR[Formula: see text], CCAAT/enhancer-binding protein [Formula: see text] (C/EBP[Formula: see text], fatty acid synthase, adipocyte fatty acid-binding protein, and lipoprotein lipase in 3T3-L1 cells. Rutin also repressed the expression of lipin1, which is an upstream regulator that controls PPAR[Formula: see text] and C/EBP[Formula: see text]. In addition, when 3T3-L1 was transfected with lipin1 siRNA to block lipin1 function, rutin did not affect the expressions of PPAR[Formula: see text] and C/EBP[Formula: see text]. These results suggest that rutin has an anti-adipogenic effect that acts through the suppression of lipin1, as well as PPAR[Formula: see text] and C/EBP[Formula: see text].
Collapse
Affiliation(s)
- Yo-Han Han
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Republic of Korea
| | - Ji-Ye Kee
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Republic of Korea
| | - Jinbong Park
- ‡ Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dae-Seung Kim
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Republic of Korea
| | - Soyoung Shin
- † Department of Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Dong-Hyun Youn
- ‡ Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - JongWook Kang
- ‡ Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Yunu Jung
- ‡ Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Young-Mi Lee
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Republic of Korea
| | - Jin-Han Park
- § Division of Biotechnology and Convergence, Republic of Korea
| | - Su-Jin Kim
- ¶ Department of Cosmeceutical Science, Daegu Hanny University, Yugok-dong, Kyungsan 38578, Republic of Korea
| | - Jae-Young Um
- ‡ Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seung-Heon Hong
- * Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Republic of Korea
| |
Collapse
|
25
|
Veratri Nigri Rhizoma et Radix (Veratrum nigrum L.) and Its Constituent Jervine Prevent Adipogenesis via Activation of the LKB1-AMPKα-ACC Axis In Vivo and In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:8674397. [PMID: 27143989 PMCID: PMC4837256 DOI: 10.1155/2016/8674397] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 11/18/2022]
Abstract
This study was performed in order to investigate the antiobese effects of the ethanolic extract of Veratri Nigri Rhizoma et Radix (VN), a herb with limited usage, due to its toxicology. An HPLC analysis identified jervine as a constituent of VN. By an Oil Red O assay and a Real-Time RT-PCR assay, VN showed higher antiadipogenic effects than jervine. In high-fat diet- (HFD-) induced obese C57BL/6J mice, VN administration suppressed body weight gain. The levels of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT-enhancer-binding protein alpha (C/EBPα), adipocyte fatty-acid-binding protein (aP2), adiponectin, resistin, and LIPIN1 were suppressed by VN, while SIRT1 was upregulated. Furthermore, VN activated phosphorylation of the liver kinase B1- (LKB1-) AMP-activated protein kinase alpha- (AMPKα-) acetyl CoA carboxylase (ACC) axis. Further investigation of cotreatment of VN with the AMPK agonist AICAR or AMPK inhibitor Compound C showed that VN can activate the phosphorylation of AMPKα in compensation to the inhibition of Compound C. In conclusion, VN shows antiobesity effects in HFD-induced obese C57BL/6J mice. In 3T3-L1 adipocytes, VN has antiadipogenic features, which is due to activating the LKB1-AMPKα-ACC axis. These results suggest that VN has a potential benefit in preventing obesity.
Collapse
|
26
|
Antiobesity Effects of Unripe Rubus coreanus Miquel and Its Constituents: An In Vitro and In Vivo Characterization of the Underlying Mechanism. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:4357656. [PMID: 26904142 PMCID: PMC4745304 DOI: 10.1155/2016/4357656] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/07/2015] [Accepted: 12/27/2015] [Indexed: 01/04/2023]
Abstract
Background. The objective of the present study was to perform a bioguided fractionation of unripe Rubus coreanus Miquel (uRC) and evaluate the lipid accumulation system involvement in its antiobesity activity as well as study the uRC mechanism of action. Results. After the fractionation, the BuOH fraction of uRC (uRCB) was the most active fraction, suppressing the differentiation of 3T3-L1 adipocytes in a dose-dependent manner. Moreover, after an oral administration for 8 weeks in HFD-induced obese mice, uRCB (10 and 50 mg/kg/day) produced a significant decrease in body weight, food efficiency ratio, adipose tissue weight and LDL-cholesterol, serum glucose, TC, and TG levels. Similarly, uRCB significantly suppressed the elevated mRNA levels of PPARγ in the adipose tissue in vivo. Next, we investigated the antiobesity effects of ellagic acid, erycibelline, 5-hydroxy-2-pyridinemethanol, m-hydroxyphenylglycine, and 4-hydroxycoumarin isolated from uRCB. Without affecting cell viability, five bioactive compounds decreased the lipid accumulation in the 3T3-L1 cells and the mRNA expression levels of key adipogenic genes such as PPARγ, C/EBPα, SREBP-1c, ACC, and FAS. Conclusion. These results suggest that uRC and its five bioactive compounds may be a useful therapeutic agent for body weight control by downregulating adipogenesis and lipogenesis.
Collapse
|
27
|
Han YH, Kee JY, Kim DS, Park J, Jeong MY, Mun JG, Park SJ, Lee JH, Um JY, Hong SH. Anti-obesity effects of Arctii Fructus (Arctium lappa) in white/brown adipocytes and high-fat diet-induced obese mice. Food Funct 2016; 7:5025-5033. [DOI: 10.1039/c6fo01170e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Arctii Fructus prevents the development of obesity through the regulation of white/brown adipocytes.
Collapse
|
28
|
Kim HL, Park J, Park H, Jung Y, Youn DH, Kang J, Jeong MY, Um JY. Platycodon grandiflorum A. De Candolle Ethanolic Extract Inhibits Adipogenic Regulators in 3T3-L1 Cells and Induces Mitochondrial Biogenesis in Primary Brown Preadipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:7721-7730. [PMID: 26244589 DOI: 10.1021/acs.jafc.5b01908] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This study was designed to evaluate the effects of Platycodon grandiflorum A. DC. ethanolic extract (PG) on obesity in brown/white preadipocytes. The effect of PG on the differentiation and mitochondrial biogenesis of brown adipocytes is still not examined. An in vivo study showed that PG induced weight loss in mice with high-fat-diet-induced obesity. PG successfully suppressed the differentiation of 3T3-L1 cells by down-regulating cellular induction of the peroxisome proliferators activated receptor γ (PPARγ), CCAAT enhancer binding protein α (C/EBPα), lipin-1, and adiponectin but increasing expression of silent mating type information regulation 2 homologue 1 (SIRT1) and the phosphorylation of AMP-activated protein kinase α (AMPKα). The effect of PG on the adipogenic factors was compared with that of its bioactive compound platycodin D. In addition, PG increased expressions of mitochondria-related genes, including uncoupling protein 1 (UCP1), peroxisome proliferator activated receptor-coactivator 1 α (PGC1α), PR domain containing 16 (PRDM16), SIRT3, nuclear respiratory factor (NRF), and cytochrome C (CytC) in primary brown adipocytes. These results indicate that PG stimulates the differentiation of brown adipocytes through modulation of mitochondria-related genes and could offer clinical benefits as a supplement to treat obesity.
Collapse
Affiliation(s)
- Hye-Lin Kim
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Hyewon Park
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Yunu Jung
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Dong-Hyun Youn
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - JongWook Kang
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Mi-Young Jeong
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| | - Jae-Young Um
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University , Seoul 130-701, Republic of Korea
| |
Collapse
|