51
|
Tian X, Gao Y, Xu Z, Lian S, Ma Y, Guo X, Hu P, Li Z, Huang C. Pharmacokinetics of mangiferin and its metabolite-Norathyriol, Part 1: Systemic evaluation of hepatic first-pass effect in vitro and in vivo. Biofactors 2016; 42:533-544. [PMID: 27130074 DOI: 10.1002/biof.1291] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/21/2016] [Indexed: 12/17/2022]
Abstract
Mangiferin (MGF), a glucoside of xanthone existing in phytomedicines and food, is increasingly attracting attention on diabetes treatment, while the underlying mechanism leading to its low oral bioavailability is unclear. Norathyriol (NTR), an active metabolite with hypoglycemic activity and its exposure after MGF dosing remains unclear. Hence, a rapid and sensitive LC-MS/MS method was established and validated to determine MGF and NTR and applied in the PK study in rats. Correspondingly, the in vitro experiments on temperature-dependent uptake, and MGF metabolism in hepatocyte and enterobacteria samples were performed. Results revealed that hepatic first-pass effect slightly contributed to the poor bioavailability of MGF, based on the MGF exposure in portal vein plasma was nearly similar to that in systemic plasma, and the MGF accumulation in the liver was limited, so was that of NTR. Correspondingly, the in vitro study revealed the MGF uptake was mainly dependent on poor passive transport, possibly leading to its limited hepatic metabolism and accumulation. Moreover, the NTR exposure remained considerably low (Cmax < 3 ng/mL, AUCNTR /AUCMGF < 3%) in plasma after single MGF dosing, corresponding to its tiny proportion (0.1%) of MGF in MGF-incubated enterobacteria samples. However, given the low generation and elimination rates of NTR, NTR might accumulate in plasma and exert effects after repeated MGF dosing, although requires further study. This work is the first systemic study on PK profiles of MGF and NTR in vitro and in vivo, which is important for the interpretation on the poor bioavailability and pharmacodynamics of MGF. © 2016 BioFactors, 42(5):533-544, 2016.
Collapse
Affiliation(s)
- Xiaoting Tian
- Modernization of traditional Chinese medicine, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yu Gao
- Modernization of traditional Chinese medicine, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhou Xu
- College of life and environmental sciences, Shanghai Normal University, Shanghai, People's Republic of China
| | - Shan Lian
- Department of pharmacy ,Harbin University of Commerce, Harbin, People's Republic of China
| | - Yuanjie Ma
- Department of pharmacy ,Harbin University of Commerce, Harbin, People's Republic of China
| | - Xiaozhen Guo
- Modernization of traditional Chinese medicine, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Pei Hu
- Modernization of traditional Chinese medicine, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhixiong Li
- Modernization of traditional Chinese medicine, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, People's Republic of China.
| | - Chenggang Huang
- Modernization of traditional Chinese medicine, Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, People's Republic of China.
| |
Collapse
|
52
|
Saha S, Sadhukhan P, Sil PC. Mangiferin: A xanthonoid with multipotent anti-inflammatory potential. Biofactors 2016; 42:459-474. [PMID: 27219011 DOI: 10.1002/biof.1292] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 01/01/2023]
Abstract
Over the last era, small molecules sourced from different plants have gained attention for their varied and long-term medicinal benefits. Their advantageous therapeutic effects in diverse pathological complications lead researchers to give an ever-increasing emphasis on them and discover their novel therapeutic potentials. Among these, the heat stable, xanthonoid group of organic molecules has gained special importance with distinctive regards to the bioactive molecule mangiferin due to its solubility in water. Mangiferin, a yellow polyphenol having C-glycosyl xanthone structure, is widely present in different edible sources like mango, and possesses numerous biological activities. Extensive research with this molecule shows its antioxidant, anti-inflammatory, antidiabetic, anticancer, antimicrobial, analgesic, and immunomodulatory properties. Thus, it provides protection against a wide range of physiological disorders. The C-glucosyl linkage and polyhydroxy groups in mangiferin's structure contribute essentially to its free radical-scavenging activity. Moreover, its ability in regulating various transcription factors like NF-κB, Nrf-2, etc. and modulating the expression of different proinflammatory signaling intermediates like tumor necrosis factor-α, COX-2, etc. contribute to its anti-inflammatory, anticancer, and antidiabetic potentials. In this comprehensive article, information has been provided about the sources, chemical structure, metabolism, and different biological activities of mangiferin with special emphasis on the underlying cellular signal transduction pathways. Insights into an in-depth assessment of mangiferin's anti-inflammatory therapeutic potential have also been discussed in detail. On an overall perspective, this review aims to stage mangiferin's diversified therapeutic applications and its emerging possibility as a promising drug in future based on its anti-inflammatory property. © 2016 BioFactors, 42(5):459-474, 2016.
Collapse
Affiliation(s)
- Sukanya Saha
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | | | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, India.
| |
Collapse
|
53
|
Fomenko EV, Chi Y. Mangiferin modulation of metabolism and metabolic syndrome. Biofactors 2016; 42:492-503. [PMID: 27534809 PMCID: PMC5077701 DOI: 10.1002/biof.1309] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/21/2016] [Accepted: 06/05/2016] [Indexed: 12/17/2022]
Abstract
The recent emergence of a worldwide epidemic of metabolic disorders, such as obesity and diabetes, demands effective strategy to develop nutraceuticals or pharmaceuticals to halt this trend. Natural products have long been and continue to be an attractive source of nutritional and pharmacological therapeutics. One such natural product is mangiferin (MGF), the predominant constituent of extracts of the mango plant Mangifera indica L. Reports on biological and pharmacological effects of MGF increased exponentially in recent years. MGF has documented antioxidant and anti-inflammatory effects. Recent studies indicate that it modulates multiple biological processes involved in metabolism of carbohydrates and lipids. MGF has been shown to improve metabolic abnormalities and disorders in animal models and humans. This review focuses on the recently reported biological and pharmacological effects of MGF on metabolism and metabolic disorders. © 2016 BioFactors, 42(5):492-503, 2016.
Collapse
Affiliation(s)
| | - Yuling Chi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
54
|
Li W, Li B, Lv J, Dong L, Zhang L, Wang T. Choline supplementation improves the lipid metabolism of intrauterine-growth-restricted pigs. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 31:686-695. [PMID: 27221247 PMCID: PMC5930279 DOI: 10.5713/ajas.15.0810] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/14/2015] [Accepted: 05/19/2016] [Indexed: 12/23/2022]
Abstract
Objective The objective of this study was to investigate the effects of dietary choline supplementation on hepatic lipid metabolism and gene expression in finishing pigs with intrauterine growth retardation (IUGR). Methods Using a 2×2 factorial design, eight normal birth weight (NBW) and eight IUGR weaned pigs were fed either a basal diet (NBW pigs fed a basal diet, NC; IUGR pigs fed a basal diet, IC) or a diet supplemented with two times more choline than the basal diet (NBW pigs fed a high-choline diet, NH; IUGR pigs fed a high-choline diet, IH) until 200 d of age. Results The results showed that the IUGR pigs had reduced body weight compared with the NBW pigs (p<0.05 from birth to d 120; p = 0.07 from d 120 to 200). Increased (p<0.05) free fatty acid (FFA) and triglyceride levels were observed in the IUGR pigs compared with the NBW pigs. Choline supplementation decreased (p<0.05) the levels of FFAs and triglycerides in the serum of the pigs. The activities of malate dehydrogenase and glucose 6-phosphate dehydrogenase were both increased (p<0.05) in the livers of the IUGR pigs. Choline supplementation decreased (p<0.05) malate dehydrogenase activity in the liver of the pigs. Gene expression of fatty acid synthase (FAS) was higher (p<0.05) in the IC group than in the other groups, and choline supplementation decreased (p<0.05) FAS and acetyl-CoA carboxylase α expression in the livers of the IUGR pigs. The expression of carnitine palmitoyl transferase 1A (CPT1A) was lower (p<0.05) in the IC group than in the other groups, and choline supplementation increased (p<0.05) the expression of CPT1A in the liver of the IUGR pigs and decreased (p<0.01) the expression of hormone-sensitive lipase in both types of pigs. The gene expression of phosphatidylethanolamine N-methyltransferase (PEMT) was higher (p<0.05) in the IC group than in the other groups, and choline supplementation significantly reduced (p<0.05) PEMT expression in the liver of the IUGR pigs. Conclusion In conclusion, the lipid metabolism was abnormal in IUGR pigs, but the IUGR pigs consuming twice the normal level of choline had improved circulating lipid parameters, which could be related to the decreased activity of nicotinamide adenine dinucleotide phosphate-generating enzymes or the altered expressions of lipid metabolism-related genes.
Collapse
Affiliation(s)
- Wei Li
- College of Animal Science and Technology, Nanjing Agricultural University (NJAU), Nanjing 210095, China
| | - Bo Li
- College of Animal Science and Technology, Nanjing Agricultural University (NJAU), Nanjing 210095, China
| | - Jiaqi Lv
- College of Animal Science and Technology, Nanjing Agricultural University (NJAU), Nanjing 210095, China
| | - Li Dong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University (NJAU), Nanjing 210095, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University (NJAU), Nanjing 210095, China
| |
Collapse
|
55
|
Gonçalves-de-Albuquerque CF, Medeiros-de-Moraes IM, Oliveira FMDJ, Burth P, Bozza PT, Castro Faria MV, Silva AR, de Castro-Faria-Neto HC. Omega-9 Oleic Acid Induces Fatty Acid Oxidation and Decreases Organ Dysfunction and Mortality in Experimental Sepsis. PLoS One 2016; 11:e0153607. [PMID: 27078880 PMCID: PMC4831806 DOI: 10.1371/journal.pone.0153607] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 03/31/2016] [Indexed: 01/18/2023] Open
Abstract
Sepsis is characterized by inflammatory and metabolic alterations, which lead to massive cytokine production, oxidative stress and organ dysfunction. In severe systemic inflammatory response syndrome, plasma non-esterified fatty acids (NEFA) are increased. Several NEFA are deleterious to cells, activate Toll-like receptors and inhibit Na+/K+-ATPase, causing lung injury. A Mediterranean diet rich in olive oil is beneficial. The main component of olive oil is omega-9 oleic acid (OA), a monounsaturated fatty acid (MUFA). We analyzed the effect of OA supplementation on sepsis. OA ameliorated clinical symptoms, increased the survival rate, prevented liver and kidney injury and decreased NEFA plasma levels in mice subjected to cecal ligation and puncture (CLP). OA did not alter food intake and weight gain but diminished reactive oxygen species (ROS) production and NEFA plasma levels. Carnitine palmitoyltransferase IA (CPT1A) mRNA levels were increased, while uncoupling protein 2 (UCP2) liver expression was enhanced in mice treated with OA. OA also inhibited the decrease in 5' AMP-activated protein kinase (AMPK) expression and increased the enzyme expression in the liver of OA-treated mice compared to septic animals. We showed that OA pretreatment decreased NEFA concentration and increased CPT1A and UCP2 and AMPK levels, decreasing ROS production. We suggest that OA has a beneficial role in sepsis by decreasing metabolic dysfunction, supporting the benefits of diets high in monounsaturated fatty acids (MUFA).
Collapse
Affiliation(s)
| | | | | | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24020–15 Niterói, RJ, Brazil
| | - Patrícia Torres Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, 21040–900 Rio de Janeiro, RJ, Brazil
| | - Mauro Velho Castro Faria
- Departamento de Medicina Interna, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, 20550–900 Rio de Janeiro, RJ, Brazil
| | - Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, 21040–900 Rio de Janeiro, RJ, Brazil
- * E-mail: (ARS); (HCCFN)
| | - Hugo Caire de Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, FIOCRUZ, 21040–900 Rio de Janeiro, RJ, Brazil
- Universidade Estácio de Sá, Programa de Produtividade Científica, Rio de Janeiro, RJ, Brazil
- * E-mail: (ARS); (HCCFN)
| |
Collapse
|
56
|
The Compound of Mangiferin-Berberine Salt Has Potent Activities in Modulating Lipid and Glucose Metabolisms in HepG2 Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8753436. [PMID: 27123455 PMCID: PMC4829696 DOI: 10.1155/2016/8753436] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/28/2016] [Accepted: 03/07/2016] [Indexed: 12/20/2022]
Abstract
The mangiferin-berberine (MB) salt was synthesized by ionic bonding of mangiferin (M) and berberine (B) at an equal molecular ratio. This study aimed to investigate the activities of MB salt in modulating lipid and glucose metabolisms in HepG2 cells. After 24 h treatment of the studying compounds, cellular AMP-activated protein kinase α (AMPKα)/acetyl-CoA carboxylase (ACC) protein levels and carnitine palmitoyltransferase (CPT) 1 activities, intracellular lipid contents, mRNA expression levels of target genes, glucose consumption, and glucose production amounts were determined. Compound C (CC) was used in the blocking experiments. Our results showed that MB salt increased p-AMPKα (Thr172)/p-ACC (Ser79) levels and CPT1 activity and suppressed oleic acid- (OA-) induced lipid accumulation and upregulation of lipogenic genes potently in HepG2 cells. The above activities of MB salt were AMPK dependent and were superior to those of M or B when administered at an equal molar concentration. MB salt enhanced basal and insulin-stimulated glucose consumption and suppressed gluconeogenesis more potently than M or B alone. The inhibiting activity of MB salt on cellular gluconeogenesis was AMPK dependent. Our results may support MB salt as a new kind of agent for the development of novel lipid or glucose-lowering drugs in the future.
Collapse
|
57
|
Gómez-Caravaca AM, López-Cobo A, Verardo V, Segura-Carretero A, Fernández-Gutiérrez A. HPLC-DAD-q-TOF-MS as a powerful platform for the determination of phenolic and other polar compounds in the edible part of mango and its by-products (peel, seed, and seed husk). Electrophoresis 2016; 37:1072-84. [DOI: 10.1002/elps.201500439] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Ana María Gómez-Caravaca
- Department of Analytical Chemistry; University of Granada; Granada Spain
- Research and Development of Functional Food Centre (CIDAF); PTS Granada; Granada Spain
| | - Ana López-Cobo
- Research and Development of Functional Food Centre (CIDAF); PTS Granada; Granada Spain
| | - Vito Verardo
- Department of Chemistry and Physics (Analytical Chemistry Area) and Research Centre for Agricultural and Food Biotechnology (BITAL), Agrifood Campus of International Excellence, ceiA3; University of Almería; Almería Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry; University of Granada; Granada Spain
- Research and Development of Functional Food Centre (CIDAF); PTS Granada; Granada Spain
| | - Alberto Fernández-Gutiérrez
- Department of Analytical Chemistry; University of Granada; Granada Spain
- Research and Development of Functional Food Centre (CIDAF); PTS Granada; Granada Spain
| |
Collapse
|
58
|
Zhou L, Pan Y, Chonan R, Batey R, Rong X, Yamahara J, Wang J, Li Y. Mitigation of Insulin Resistance by Mangiferin in a Rat Model of Fructose-Induced Metabolic Syndrome Is Associated with Modulation of CD36 Redistribution in the Skeletal Muscle. J Pharmacol Exp Ther 2016; 356:74-84. [PMID: 26498906 DOI: 10.1124/jpet.115.229005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/22/2015] [Indexed: 11/22/2022] Open
Abstract
Mangiferin is one of the prominent active components responsible for the antidiabetic property of many traditional herbs, but its underlying mechanisms of action remain unclear. CD36 in skeletal muscle is known to contribute to the etiology of insulin resistance by facilitating fatty acid uptake. This study investigated the effect of mangiferin on insulin resistance. The results showed that treatment of Wistar-Kyoto rats with mangiferin (15 mg/kg, once daily, by oral gavage) for 7 weeks inhibited chronic liquid fructose consumption-induced increases in plasma insulin concentrations at the baseline and during oral glucose tolerance test (OGTT), and the homeostasis model assessment of insulin resistance index. It also suppressed the increases in fasted plasma nonesterified fatty acid (NEFA) concentration and the adipose tissue insulin resistance index. Mechanistically, mangiferin neither affected intakes of fructose and chow, and the increase in epididymal and perirenal fat, nor attenuated fructose-induced hypertension. In contrast, mangiferin attenuated fructose-induced acceleration of plasma NEFA clearance during OGTT, and tended to decrease excessive triglyceride accumulation in gastrocnemius. Immunofluorescence staining and subsequent rating of CD36-expressing fibers in gastrocnemius revealed that mangiferin restored fructose-stimulated sarcolemmal CD36 overexpression and decreased intracellular CD36 distribution. In addition, the effects of mangiferin on the parameters associated with insulin resistance and abnormal fatty acid metabolism were absent in the spontaneously hypertensive rats carrying numerous nonfunctional mutations in the CD36 gene. Thus, these results suggest that mangiferin treatment mitigates insulin resistance in a rat model of fructose-induced metabolic syndrome by modulating sarcolemmal and intracellular CD36 redistribution in the skeletal muscle.
Collapse
Affiliation(s)
- Liang Zhou
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Yongquan Pan
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Ritsu Chonan
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Robert Batey
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Xianglu Rong
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Johji Yamahara
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Jianwei Wang
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| | - Yuhao Li
- Faculty of Basic Medical Sciences (L.Z.), Laboratory Animal Center (Y.P.), and Laboratory of Traditional Chinese Medicine (J.W.), Chongqing Medical University, Chongqing, China; Koei Kogyo Co., Ltd., Tokyo, Japan (R.C.); Central Clinical School, Royal Prince Alfred Hospital, The University of Sydney, Sydney, Australia (R.B.); Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, and the Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China (X.R.); Pharmafood Institute, Kyoto, Japan (J.Y.); and Endocrinology and Metabolism Group, Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, Australia (Y.L.)
| |
Collapse
|
59
|
Grahame Hardie D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm Sin B 2016; 6:1-19. [PMID: 26904394 PMCID: PMC4724661 DOI: 10.1016/j.apsb.2015.06.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022] Open
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that is almost universally expressed in eukaryotic cells. While it appears to have evolved in single-celled eukaryotes to regulate energy balance in a cell-autonomous manner, during the evolution of multicellular animals its role has become adapted so that it also regulates energy balance at the whole body level, by responding to hormones that act primarily on the hypothalamus. AMPK monitors energy balance at the cellular level by sensing the ratios of AMP/ATP and ADP/ATP, and recent structural analyses of the AMPK heterotrimer that have provided insight into the complex mechanisms for these effects will be discussed. Given the central importance of energy balance in diseases that are major causes of morbidity or death in humans, such as type 2 diabetes, cancer and inflammatory disorders, there has been a major drive to develop pharmacological activators of AMPK. Many such activators have been described, and the various mechanisms by which these activate AMPK will be discussed. A particularly large class of AMPK activators are natural products of plants derived from traditional herbal medicines. While the mechanism by which most of these activate AMPK has not yet been addressed, I will argue that many of them may be defensive compounds produced by plants to deter infection by pathogens or grazing by insects or herbivores, and that many of them will turn out to be inhibitors of mitochondrial function.
Collapse
Affiliation(s)
- David Grahame Hardie
- Division of Cell Signaling & Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| |
Collapse
|
60
|
Ganji SH, Kashyap ML, Kamanna VS. Niacin inhibits fat accumulation, oxidative stress, and inflammatory cytokine IL-8 in cultured hepatocytes: Impact on non-alcoholic fatty liver disease. Metabolism 2015; 64:982-90. [PMID: 26024755 DOI: 10.1016/j.metabol.2015.05.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 04/06/2015] [Accepted: 05/04/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is a common disorder characterized by excessive hepatic fat accumulation, production of reactive oxygen species (ROS), inflammation and potentially resulting in non-alcoholic steatohepatitis (NASH), cirrhosis and end-stage liver disease. Recently, we have shown that niacin significantly prevented hepatic steatosis and regressed pre-existing steatosis in high-fat fed rat model of NAFLD. To gain further insight into the cellular mechanisms, this study investigated the effect of niacin on human hepatocyte fat accumulation, ROS production, and inflammatory mediator IL-8 secretion. MATERIALS AND METHODS Human hepatoblastoma cell line HepG2 or human primary hepatocytes were first stimulated with palmitic acid followed by treatment with niacin or control for 24 h. RESULTS The data indicated that niacin (at 0.25 and 0.5 mmol/L doses) significantly inhibited palmitic acid-induced fat accumulation in human hepatocytes by 45-62%. This effect was associated with inhibition of diacylglycerol acyltransferase 2 (DGAT2) mRNA expression without affecting the mRNA expression of fatty acid synthase (FAS) and carnitine palmitoyltransferase 1 (CPT1). Niacin attenuated hepatocyte ROS production and it also inhibited NADPH oxidase activity. Niacin reduced palmitic acid-induced IL-8 levels. CONCLUSIONS These findings suggest that niacin, through inhibiting hepatocyte DGAT2 and NADPH oxidase activity, attenuates hepatic fat accumulation and ROS production respectively. Decreased ROS production, at least in part, may have contributed to the inhibition of pro-inflammatory IL-8 levels. These mechanistic studies may be useful for the clinical development of niacin and niacin-related compounds for the treatment of NAFLD/NASH and its complications.
Collapse
Affiliation(s)
- Shobha H Ganji
- Department of Veterans Affairs Healthcare System, Long Beach, CA; University of California, Irvine, CA
| | - Moti L Kashyap
- Department of Veterans Affairs Healthcare System, Long Beach, CA; University of California, Irvine, CA
| | - Vaijinath S Kamanna
- Department of Veterans Affairs Healthcare System, Long Beach, CA; University of California, Irvine, CA.
| |
Collapse
|
61
|
Han J, Yang N, Zhang F, Zhang C, Liang F, Xie W, Chen W. Rhizoma Anemarrhenae extract ameliorates hyperglycemia and insulin resistance via activation of AMP-activated protein kinase in diabetic rodents. JOURNAL OF ETHNOPHARMACOLOGY 2015; 172:368-376. [PMID: 26162543 DOI: 10.1016/j.jep.2015.05.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 05/04/2015] [Accepted: 05/07/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhizoma Anemarrhenae has been used in Asian countries for thousands of years to treat diabetes. Insulin resistance (IR) is the primary cause responsible for type 2 diabetes. The aim of this study was to to assess the hypoglycemic and insulin sensitizing properties of Rhizoma Anemarrhenae extract (TFA) in animal models of insulin resistance and/or diabetes and to delineate modes of action. MATERIALS AND METHODS In-vivo studies were performed on STZ-induced diabetic mice and KK-Ay mice, the former of which were given the extract alone or in combination with insulin for 7 days, and the latter of which were given the extract for 8 consecutive weeks. Fasting blood glucose and serum insulin levels were measured. Pancreatic tissue sections were examined using transmission electron micrographs. Further, hyperinsulinemic-euglycemic clamping study was conducted in BCG vaccine-induced insulin resistance rats, and glucose infusion rate was examined. Mechanisms of action were investigated in 3T3-L1 and Hela cells using Western blot analysis. RESULTS Our study showed that TFA enhanced the glucose-lowering effects of exogenous insulin administration in STZ-induced diabetic mice. Therapeutic administration of TFA significantly reduced fasting blood glucose, and serum insulin levels, and markedly increased the size and the number of insulin-producing beta cells in KK-Ay mice. Further, hyperinsulinemic-euglycemic clamping study showed that glucose infusion rate was significantly improved in TFA-treated BCG vaccine-induced insulin resistance rats. Study of mechanism of action revealed that TFA increased phosphorylation of AMPK and its downstream target, acetyl-CoA carboxylase (ACC) in 3T3-L1 cells. It activates AMPK in a LKB1-independent manner, providing a unified explanation for the beneficial effects of TFA. CONCLUSIONS This study that TFA mediates activation of AMPK and improves overall glucose and lipid metabolism in diabetic rodents, highlights the potential utility of TFA for the management of type 2 diabetes.
Collapse
Affiliation(s)
- Jun Han
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China
| | - Na Yang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China
| | - Feng Zhang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China
| | - Chuan Zhang
- New Drug Research Center, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, PR China
| | - Fengying Liang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China
| | - WeiFen Xie
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China
| | - Wansheng Chen
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, PR China.
| |
Collapse
|
62
|
Chu X, He X, Shi Z, Li C, Guo F, Li S, Li Y, Na L, Sun C. Ursolic acid increases energy expenditure through enhancing free fatty acid uptake and β-oxidation via an UCP3/AMPK-dependent pathway in skeletal muscle. Mol Nutr Food Res 2015; 59:1491-503. [PMID: 25944715 DOI: 10.1002/mnfr.201400670] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 04/16/2015] [Accepted: 04/21/2015] [Indexed: 12/12/2022]
Abstract
SCOPE Ursolic acid (UA) is a triterpenoid compound with multifold biological functions. Our previous studies have reported that UA protects against high-fat diet-induced obesity and improves insulin resistance (IR). However, the potential mechanisms are still undefined. Free fatty acid (FFA) metabolism in skeletal muscle plays a central role in obesity and IR. Therefore, in this study, we investigated the effect and the potential mechanisms of UA on skeletal muscle FFA metabolism. METHODS AND RESULTS In diet-induced obese rats, 0.5% UA supplementation for 6 weeks markedly reduced body weight, increased energy expenditure, decreased FFA level in serum and skeletal muscle and triglyceride content in skeletal muscle. In vitro, the data provided directly evidence that UA significantly increased fluorescently labeled FFA uptake and (3) H-labeled palmitic acid β-oxidation. UA-activated AMP-activated protein kinase (AMPK) and downstream targets were involved in the increase of FFA catabolism. Moreover, upregulated uncoupling protein 3 (UCP3) by UA contributed to AMPK activation via elevating adenosine monophosphate/adenosine triphosphate ratio. CONCLUSION UA increases FFA burning through enhancing skeletal muscle FFA uptake and β-oxidation via an UCP3/AMPK-dependent pathway, which provides a novel perspective on the biological function of UA against obesity and IR.
Collapse
Affiliation(s)
- Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Xuan He
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Zhiping Shi
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Chunjuan Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Fuchuan Guo
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
- Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Lixin Na
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
- Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| |
Collapse
|
63
|
Zhang S, Chen G, Li N, Dai M, Chen C, Wang P, Tang H, Hoopes SL, Zeldin DC, Wang DW, Xu X. CYP2J2 overexpression ameliorates hyperlipidemia via increased fatty acid oxidation mediated by the AMPK pathway. Obesity (Silver Spring) 2015; 23:1401-13. [PMID: 26053032 PMCID: PMC4565055 DOI: 10.1002/oby.21115] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 03/16/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The study aims to investigate the effect of cytochrome P450 2J2 (CYP2J2) overexpression on hyperlipidemia in mice and further to explore its effect on fatty acid oxidation in vivo and in vitro. METHODS The effects and mechanisms of endothelial-specific CYP2J2 transgene (Tie2-CYP2J2-Tr) on lipid and fatty acid metabolism were investigated in high-fat diet (HFD) -treated mice. HepG2, LO2 cells, and HUVECs were exposed to 0.4 mM free fatty acid (FFA) for 24 h and used as a model to investigate the roles of CYP2J2 overexpression and epoxyeicosatrienoic acids (EETs) on fatty acid β-oxidation in vitro. RESULTS Tie2-CYP2J2-Tr mice had significantly lower plasma and liver triglycerides, lower liver cholesterol and fatty acids, and reduced HFD-induced lipid accumulation. CYP2J2 overexpression resulted in activation of the hepatic and endothelial AMPKα, increased ACC phosphorylation, and increased expression of CPT-1 and PPARα, which were all reduced by HFD treatment. In FFA-treated HepG2, LO2, and HUVECs, both CYP2J2 overexpression and EETs significantly decreased lipid accumulation and increased fatty acid oxidation via activating the AMPK and PPARα pathways. CONCLUSIONS Endothelial-specific CYP2J2 overexpression alleviates HFD-induced hyperlipidemia in vivo. CYP2J2 ameliorates FFA-induced dyslipidemia via increased fatty acid oxidation mediated by the AMPK and PPARα pathways.
Collapse
Affiliation(s)
- Shasha Zhang
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - Guangzhi Chen
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - Ning Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, University of Chinese Academy of Sciences, Wuhan 430071, People’s Republic of China
| | - Meiyan Dai
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - Chen Chen
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - Peihua Wang
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - Huiru Tang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Centre for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, University of Chinese Academy of Sciences, Wuhan 430071, People’s Republic of China
| | - Samantha L. Hoopes
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Dao Wen Wang
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| | - Xizhen Xu
- Departments of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030
| |
Collapse
|
64
|
Mangiferin supplementation improves serum lipid profiles in overweight patients with hyperlipidemia: a double-blind randomized controlled trial. Sci Rep 2015; 5:10344. [PMID: 25989216 PMCID: PMC4437311 DOI: 10.1038/srep10344] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/10/2015] [Indexed: 12/21/2022] Open
Abstract
Our previous studies have shown that mangiferin decreased serum triglycerides and free fatty acids (FFAs) by increasing FFAs oxidation in both animal and cell experiments. This study sought to evaluate the effects of mangiferin on serum lipid profiles in overweight patients with hyperlipidemia. Overweight patients with hyperlipidemia (serum triglyceride ≥ 1.70 mmol/L, and total cholesterol ≥ 5.2 mmol/L) were included in this double-blind randomized controlled trial. Participants were randomly allocated to groups, either receiving mangiferin (150 mg/day) or identical placebo for 12 weeks. The lipid profile and serum levels of mangiferin, glucose, L-carnitine, β-hydroxybutyrate, and acetoacetate were determined at baseline and 12 weeks. A total of 97 participants completed the trial. Compared with the placebo control, mangiferin supplementation significantly decreased the serum levels of triglycerides and FFAs, and insulin resistance index. Mangiferin supplementation also significantly increased the serum levels of mangiferin, high-density lipoprotein cholesterol, L-carnitine, β-hydroxybutyrate, and acetoacetate, and increased lipoprotein lipase activity. However, there were no differences in the serum levels of total cholesterol, low-density lipoprotein cholesterol, serum glucose, and insulin between groups. Mangiferin supplementation could improve serum lipid profiles by reducing serum triglycerides and FFAs in overweight patients with hyperlipidemia, partly due to the promotion of FFAs oxidation.
Collapse
|
65
|
Han J, Yi J, Liang F, Jiang B, Xiao Y, Gao S, Yang N, Hu H, Xie WF, Chen W. X-3, a mangiferin derivative, stimulates AMP-activated protein kinase and reduces hyperglycemia and obesity in db/db mice. Mol Cell Endocrinol 2015; 405:63-73. [PMID: 25681564 DOI: 10.1016/j.mce.2015.02.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/12/2015] [Accepted: 02/06/2015] [Indexed: 12/19/2022]
Abstract
Diabetes mellitus is a major health concern, affecting nearly 10% of the population. Here we describe a potential novel therapeutic agent for this disease, X-3, a derivative of mangiferin. Therapeutic administration of X-3 significantly and dose-dependently reduced plasma glucose and triglycerides in db/db mice following 8 week-treatments. Treatment with X-3 dose-dependently increased the number of insulin-positive β-cell mass. Importantly, X-3 did not cause any death or signs of toxicity in acute toxicity studies. Study of mechanism of action revealed that X-3 increased glucose uptake in parallel with increased phosphorylation of AMP-activated protein kinase (AMPK) in 3T3-L1 cells. It activates AMPK in both LKB1-dependent and -independent manner. Furthermore, administration of X-3 resulted in activation of AMPK and its downstream target, acetyl-CoA carboxylase (ACC) in the hypothalamus, liver, muscle and adipose tissues of C57BL/6 mice. An 80 mg/kg X-3 was more potent than metformin at 500 mg/kg in the hypothalamus, and interscapular fat tissues, potent than MF at the same dose in the liver. Thus, we conclude that X-3 is a promising new class of AMPK activating drug, and can potentially be used in the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Jun Han
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jia Yi
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Fengying Liang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bo Jiang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ying Xiao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Shouhong Gao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Na Yang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Honggang Hu
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wansheng Chen
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
66
|
Boone-Villa D, Aguilera-Méndez A, Miranda-Cervantes A, Fernandez-Mejia C. Effects of Biotin Supplementation in the Diet on Adipose Tissue cGMP Concentrations, AMPK Activation, Lipolysis, and Serum-Free Fatty Acid Levels. J Med Food 2015; 18:1150-6. [PMID: 25835526 DOI: 10.1089/jmf.2014.0170] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Several studies have shown that pharmacological concentrations of biotin decrease hyperlipidemia. The molecular mechanisms by which pharmacological concentrations of biotin modify lipid metabolism are largely unknown. Adipose tissue plays a central role in lipid homeostasis. In the present study, we analyzed the effects of biotin supplementation in adipose tissue on signaling pathways and critical proteins that regulate lipid metabolism, as well as on lipolysis. In addition, we assessed serum fatty acid concentrations. Male BALB/cAnN Hsd mice were fed a control or a biotin-supplemented diet (control: 1.76 mg biotin/kg; supplemented: 97.7 mg biotin/kg diet) over 8 weeks postweaning. Compared with the control group, biotin-supplemented mice showed an increase in the levels of adipose guanosine 3',5'-cyclic monophosphate (cGMP) (control: 30.3±3.27 pmol/g wet tissue; supplemented: 49.5±3.44 pmol/g wet tissue) and of phosphorylated forms of adenosine 5'-monophosphate-activated protein kinase (AMPK; 65.2%±1.06%), acetyl-coenzyme A (CoA), carboxylase-1 (196%±68%), and acetyl-CoA carboxylase-2 (78.1%±18%). Serum fatty acid concentrations were decreased (control: 1.12±0.04 mM; supplemented: 0.91±0.03 mM), and no change in lipolysis was found (control: 0.29±0.05 μmol/mL; supplemented: 0.33±0.08 μmol/mL). In conclusion, 8 weeks of dietary biotin supplementation increased adipose tissue cGMP content and protein expression of the active form of AMPK and of the inactive forms of acetyl-CoA carboxylase-1 and acetyl-CoA carboxylase-2. Serum fatty acid levels fell, and no change in lipolysis was observed. These findings provide insight into the effects of biotin supplementation on adipose tissue and support its use in the treatment of dyslipidemia.
Collapse
Affiliation(s)
- Daniel Boone-Villa
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Asdrubal Aguilera-Méndez
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Adriana Miranda-Cervantes
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Cristina Fernandez-Mejia
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Instituto Nacional de Pediatría, Mexico City, Mexico
| |
Collapse
|
67
|
Seo MS, Kim JH, Kim HJ, Chang KC, Park SW. Honokiol activates the LKB1–AMPK signaling pathway and attenuates the lipid accumulation in hepatocytes. Toxicol Appl Pharmacol 2015; 284:113-24. [DOI: 10.1016/j.taap.2015.02.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 02/14/2015] [Accepted: 02/21/2015] [Indexed: 02/06/2023]
|
68
|
Song J, Li J, Hou F, Wang X, Liu B. Mangiferin inhibits endoplasmic reticulum stress-associated thioredoxin-interacting protein/NLRP3 inflammasome activation with regulation of AMPK in endothelial cells. Metabolism 2015; 64:428-37. [PMID: 25499441 DOI: 10.1016/j.metabol.2014.11.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/15/2014] [Accepted: 11/25/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Endothelial dysfunction is tightly associated with cardiovascular complications in diabetic patients. This study aims to investigate the effects of mangiferin on the regulation of endothelial homeostasis under endoplasmic reticulum stress (ER stress) conditions. RESULTS High glucose (25 mmol/L) exposure induced ER stress and promoted ROS production in endothelial cells. Mangiferin effectively inhibited ER stress-associated oxidative stress by attenuating IRE1α phosphorylation and reducing ROS production. In response to ER stress, thioredoxin-interacting protein (TXNIP) expression increased, followed by NLRP3 inflammasome activation and increased IL-1β secretion. Mangiferin treatment attenuated the expressions of TXNIP and NLRP3 and reduced IL-1β and IL-6 production, demonstrating its inhibitory effects on TXNIP/NLRP3 inflammasome activation. NLRP3 inflammasome activation is responsible for mitochondrial cell death. Mangiferin restored the loss of the mitochondrial membrane potential (Δψm) and inhibited caspase-3 activity, and thereby protected cells from high glucose-induced apoptosis. Moreover, mangiferin inhibited ET-1 secretion and restored the loss of NO production when cells were exposed to high glucose. Mangiferin enhanced AMPK phosphorylation and AMPK inhibitor compound C diminished its beneficial effects, indicating the potential role of AMPK in its action. CONCLUSION Our work showed the beneficial effects of mangiferin on the improvement of endothelial homeostasis and elucidated the molecular pathway through which mangiferin ameliorated endothelial dysfunction by inhibition of ER stress-associated TXNIP/NLRP3 inflammasome activation in endothelial cells. SIGNIFICANCE These findings demonstrated the beneficial effects of mangiferin on the regulation of endothelial homeostasis and indicated its potential application in the management of diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Junna Song
- Hebei University of Chinese Medicine College of pharmacy teaching and researching section of medicinal plant, Hebei, China
| | - Jia Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicines, Nanjing, China
| | - Fangjie Hou
- Hebei University of Chinese Medicine College of pharmacy teaching and researching section of medicinal plant, Hebei, China
| | - Xiaona Wang
- Hebei University of Chinese Medicine College of pharmacy teaching and researching section of medicinal plant, Hebei, China
| | - Baolin Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicines, Nanjing, China.
| |
Collapse
|
69
|
Beneficial effects of neomangiferin on high fat diet-induced nonalcoholic fatty liver disease in rats. Int Immunopharmacol 2015; 25:218-28. [PMID: 25661699 DOI: 10.1016/j.intimp.2015.01.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/12/2015] [Accepted: 01/28/2015] [Indexed: 02/08/2023]
Abstract
This study was carried out to determine the effect and mechanism of action of neomangiferin (NG) on high-fat diet-induced nonalcoholic fatty liver disease (NAFLD) in rats. NAFLD rats were randomly assigned into several groups of equal number. NG (50, 25mg/kg·day(-1) BW) and lipanthyl (PT, 5mg/kg·day(-1) BW) were given to the NAFLD rats, respectively. In the study, serum lipids, metabolic rate, liver fat, liver lipids and histology were examined. To further investigate the molecular mechanism of the effect of NG on NAFLD, expression levels of mRNA and protein for peroxisome proliferator-activated receptor α (PPARα), fatty acid transport protein 2 (FATP2), long-chain-fatty-acid - CoA ligase 1 (ACSL1) and carnitine palmitoyltransferase 1a (CPT1a) in the liver were determined by Real Time-PCR and western blot analysis, respectively. NG administration significantly reduced the final body weight, liver fat accumulation, and serum triglyceride (TG), total cholesterol (TC) concentrations, low-density lipoprotein cholesterol (LDL-C), glucose (GLU) levels, and hepatic TG, TC, malondialdehyde (MDA) levels, but increased serum high-density lipoprotein cholesterol (HDL-C) and hepatic superoxide dismutase (SOD) levels. NG upregulated the mRNA and protein expression of PPARα and CPT1a, but downregulated the mRNA and protein expression of FATP2 and ACSL1 in the liver. These results suggested that NG can regulate NAFLD partly by modulating the expression levels of genes involved in FFA uptake and lipid oxidation.
Collapse
|
70
|
Zhou C, Li G, Li Y, Gong L, Huang Y, Shi Z, Du S, Li Y, Wang M, Yin J, Sun C. A high-throughput metabolomic approach to explore the regulatory effect of mangiferin on metabolic network disturbances of hyperlipidemia rats. MOLECULAR BIOSYSTEMS 2015; 11:418-33. [DOI: 10.1039/c4mb00421c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This paper was designed to study metabolomic characters of the high-fat diet (HFD)-induced hyperlipidemia and the intervention effects of Mangiferin (MG).
Collapse
|
71
|
Seo MS, Hong SW, Yeon SH, Kim YM, Um KA, Kim JH, Kim HJ, Chang KC, Park SW. Magnolia officinalis attenuates free fatty acid-induced lipogenesis via AMPK phosphorylation in hepatocytes. JOURNAL OF ETHNOPHARMACOLOGY 2014; 157:140-148. [PMID: 25261688 DOI: 10.1016/j.jep.2014.09.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/21/2014] [Accepted: 09/17/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnolia officinalis (MO) is a traditional Chinese herbal medicine that has been used in clinical practice to treat liver disease. The aim of this study is to examine the effects of MO on the development of nonalcoholic fatty liver in hepatocytes. MATERIALS AND METHODS Human hepatoma-derived HepG2 cells and mouse normal FL83B hepatocytes were exposed to 0.5mM free fatty acids (FFAs; oleate:palmitate, 2:1) for 24h to simulate conditions of nonalcoholic fatty liver in vitro. The cells were treated with a standardized MO extract 1h prior to FFA exposure. RESULTS MO pretreatment attenuated the increases in intracellular lipid accumulation and triglyceride content in FFA-exposed hepatocytes in a dose-dependent manner. MO pretreatment significantly inhibited both sterol regulatory element-binding protein (SREBP)-1c activation and increases in fatty acid translocase, fatty acid synthase, and stearoyl CoA desaturase-1 protein expression in FFA-exposed hepatocytes in a dose-dependent manner. MO pretreatment markedly induced adenosine monophosphate-activated protein kinase (AMPK) phosphorylation in hepatocytes. Compound C, an AMPK inhibitor, blocked the inhibitory effect of MO on the increases in intracellular lipid accumulation and triglyceride content induced by FFAs. In hepatocytes pretreated with compound C, MO failed to inhibit SREBP-1c activation and the increases in fatty acid translocase, fatty acid synthase, and stearoyl-CoA desaturase-1 protein expression induced by FFAs. CONCLUSIONS Our results indicate that MO attenuates triglyceride biosynthesis and accumulation induced by FFAs in hepatocytes, suggesting its pharmacological potential for the prevention of nonalcoholic fatty liver disease. These effects may be mediated by the inhibition of SREBP-1c via AMPK phosphorylation.
Collapse
Affiliation(s)
- Min Suk Seo
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, 816-15 Jinjudaero, Jinju 660-751, Republic of Korea
| | - Sung-Woon Hong
- R&D Center, Huons Co. Ltd., Ansan 426-791, Republic of Korea
| | - Sung Hum Yeon
- R&D Center, Huons Co. Ltd., Ansan 426-791, Republic of Korea
| | - Young-Mok Kim
- R&D Center, Huons Co. Ltd., Ansan 426-791, Republic of Korea
| | - Key An Um
- R&D Center, Huons Co. Ltd., Ansan 426-791, Republic of Korea
| | - Jung Hwan Kim
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, 816-15 Jinjudaero, Jinju 660-751, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, 816-15 Jinjudaero, Jinju 660-751, Republic of Korea
| | - Ki Churl Chang
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, 816-15 Jinjudaero, Jinju 660-751, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, 816-15 Jinjudaero, Jinju 660-751, Republic of Korea.
| |
Collapse
|
72
|
Apontes P, Liu Z, Su K, Benard O, Youn DY, Li X, Li W, Mirza RH, Bastie CC, Jelicks LA, Pessin JE, Muzumdar RH, Sauve AA, Chi Y. Mangiferin stimulates carbohydrate oxidation and protects against metabolic disorders induced by high-fat diets. Diabetes 2014; 63:3626-36. [PMID: 24848064 PMCID: PMC4207399 DOI: 10.2337/db14-0006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Excessive dietary fat intake causes systemic metabolic toxicity, manifested in weight gain, hyperglycemia, and insulin resistance. In addition, carbohydrate utilization as a fuel is substantially inhibited. Correction or reversal of these effects during high-fat diet (HFD) intake is of exceptional interest in light of widespread occurrence of diet-associated metabolic disorders in global human populations. Here we report that mangiferin (MGF), a natural compound (the predominant constituent of Mangifera indica extract from the plant that produces mango), protected against HFD-induced weight gain, increased aerobic mitochondrial capacity and thermogenesis, and improved glucose and insulin profiles. To obtain mechanistic insight into the basis for these effects, we determined that mice exposed to an HFD combined with MGF exhibited a substantial shift in respiratory quotient from fatty acid toward carbohydrate utilization. MGF treatment significantly increased glucose oxidation in muscle of HFD-fed mice without changing fatty acid oxidation. These results indicate that MGF redirects fuel utilization toward carbohydrates. In cultured C2C12 myotubes, MGF increased glucose and pyruvate oxidation and ATP production without affecting fatty acid oxidation, confirming in vivo and ex vivo effects. Furthermore, MGF inhibited anaerobic metabolism of pyruvate to lactate but enhanced pyruvate oxidation. A key target of MGF appears to be pyruvate dehydrogenase, determined to be activated by MGF in a variety of assays. These findings underscore the therapeutic potential of activation of carbohydrate utilization in correction of metabolic syndrome and highlight the potential of MGF to serve as a model compound that can elicit fuel-switching effects.
Collapse
Affiliation(s)
- Pasha Apontes
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Zhongbo Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Kai Su
- Department of Paediatrics, Albert Einstein College of Medicine, Bronx, NY
| | | | - Dou Y Youn
- Department of Pharmacology, Weill Cornell Medical College, New York, NY
| | - Xisong Li
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Wei Li
- Department of Pharmacology, Weill Cornell Medical College, New York, NY
| | - Raihan H Mirza
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Claire C Bastie
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Linda A Jelicks
- Department of Physiology & Biophysics and Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY
| | - Radhika H Muzumdar
- Department of Paediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medical College, New York, NY
| | - Yuling Chi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
73
|
Mahali SK, Verma N, Manna SK. Advanced Glycation End Products Induce Lipogenesis: Regulation by Natural Xanthone through Inhibition of ERK and NF-κB. J Cell Physiol 2014; 229:1972-80. [DOI: 10.1002/jcp.24647] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/11/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Sidhartha K. Mahali
- Laboratory of Immunology; Centre for DNA Fingerprinting and Diagnostics; Nampally Hyderabad India
| | - Neeharika Verma
- Laboratory of Immunology; Centre for DNA Fingerprinting and Diagnostics; Nampally Hyderabad India
| | - Sunil K. Manna
- Laboratory of Immunology; Centre for DNA Fingerprinting and Diagnostics; Nampally Hyderabad India
| |
Collapse
|
74
|
Xing X, Li D, Chen D, Zhou L, Chonan R, Yamahara J, Wang J, Li Y. Mangiferin treatment inhibits hepatic expression of acyl-coenzyme A:diacylglycerol acyltransferase-2 in fructose-fed spontaneously hypertensive rats: a link to amelioration of fatty liver. Toxicol Appl Pharmacol 2014; 280:207-15. [PMID: 25123789 DOI: 10.1016/j.taap.2014.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 12/20/2022]
Abstract
Mangiferin, a xanthone glucoside, and its associated traditional herbs have been demonstrated to improve abnormalities of lipid metabolism. However, its underlying mechanisms remain largely unclear. This study investigated the anti-steatotic effect of mangiferin in fructose-fed spontaneously hypertensive rat (SHR)s that have a mutation in sterol regulatory element binding protein (SREBP)-1. The results showed that co-administration of mangiferin (15 mg/kg, once daily, by oral gavage) over 7 weeks dramatically diminished fructose-induced increases in hepatic triglyceride content and Oil Red O-stained area in SHRs. However, blood pressure, fructose and chow intakes, white adipose tissue weight and metabolic parameters (plasma concentrations of glucose, insulin, triglyceride, total cholesterol and non-esterified fatty acids) were unaffected by mangiferin treatment. Mechanistically, mangiferin treatment suppressed acyl-coenzyme A:diacylglycerol acyltransferase (DGAT)-2 expression at the mRNA and protein levels in the liver. In contrast, mangiferin treatment was without effect on hepatic mRNA and/or protein expression of SREBP-1/1c, carbohydrate response element binding protein, liver pyruvate kinase, fatty acid synthase, acetyl-CoA carboxylase-1, stearoyl-CoA desaturase-1, DGAT-1, monoacyglycerol acyltransferase-2, microsomal triglyceride transfer protein, peroxisome proliferator-activated receptor-alpha, carnitine palmitoyltransferase-1 and acyl-CoA oxidase. Collectively, our results suggest that mangiferin treatment ameliorates fatty liver in fructose-fed SHRs by inhibiting hepatic DGAT-2 that catalyzes the final step in triglyceride biosynthesis. The anti-steatotic effect of mangiferin may occur independently of the hepatic signals associated with de novo fatty acid synthesis and oxidation.
Collapse
Affiliation(s)
- Xiaomang Xing
- Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016 China
| | - Danyang Li
- Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016 China
| | - Dilong Chen
- Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016 China
| | - Liang Zhou
- Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016 China
| | | | | | - Jianwei Wang
- Department of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016 China.
| | - Yuhao Li
- Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, NSW 2000 Australia.
| |
Collapse
|
75
|
Shimada T, Nakayama Y, Harasawa Y, Matsui H, Kobayashi H, Sai Y, Miyamoto KI, Tomatsu S, Aburada M. Salacia reticulata has therapeutic effects on obesity. J Nat Med 2014; 68:668-76. [DOI: 10.1007/s11418-014-0845-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/27/2014] [Indexed: 02/06/2023]
|
76
|
Abstract
The adenosine monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway arose early during evolution of eukaryotic cells, when it appears to have been involved in the response to glucose starvation and perhaps also in monitoring the output of the newly acquired mitochondria. Due to the advent of hormonal regulation of glucose homeostasis, glucose starvation is a less frequent event for mammalian cells than for single-celled eukaryotes. Nevertheless, the AMPK system has been preserved in mammals where, by monitoring cellular AMP:adenosine triphosphate (ATP) and adenosine diphosphate (ADP):ATP ratios and balancing the rates of catabolism and ATP consumption, it maintains energy homeostasis at a cell-autonomous level. In addition, hormones involved in maintaining energy balance at the whole-body level interact with AMPK in the hypothalamus. AMPK is activated by two widely used clinical drugs, metformin and aspirin, and also by many natural products of plants that are either derived from traditional medicines or are promoted as "nutraceuticals."
Collapse
Affiliation(s)
- D Grahame Hardie
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, Scotland, United Kingdom;
| |
Collapse
|
77
|
Nair SV, Zhang J, Wang Y. Ethanol extract of Liuwei Dihuang reduces weight gain and visceral fat in obese-prone CD rats fed a high-fat diet. Exp Biol Med (Maywood) 2014; 239:552-8. [PMID: 24603076 DOI: 10.1177/1535370214525313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The current study investigated the effect and mechanisms of action of Liuwei Dihuang ethanol extract (LWDH-EE) on obesity and related metabolic phenotypes in male obese-prone CD rats. The rats were fed a high-fat diet and treated with 0 (obese control), 350 (EE350), or 700 (EE700) mg/kg/d of LWDH-EE in water once a day by gavage feeding for 10 weeks. The EE700 decreased body weight after 3 weeks of the treatment and the effect was maintained throughout the remaining study period. The EE700 also significantly reduced visceral fat and improved metabolic phenotypes by lowering the serum total cholesterol (T-C), non-high-density lipoprotein cholesterol, triacylglycerol, free fatty acids (FFA), and leptin levels. The EE350 reduced epididymal fat, serum T-C, and FFA but did not significantly affect other parameters. LWDH-EE dose-dependently increased fat and carbohydrate oxidations, energy expenditure, and the relative efficiency of fat oxidation for energy expenditure. EE350 and EE700 reduced food intake only in week 5 and did not affect the accumulative food intake in every week and the entire treatment period. Taken together, the results suggest that LWDH-EE is a potential therapeutic agent for the prevention of obesity possibly through a primary action of increasing energy metabolism and expenditure, along with a possible effect of decreasing energy intake.
Collapse
Affiliation(s)
- Sandhya Vg Nair
- Aquatic and Crop Resource Development, National Research Council of Canada, Charlottetown, PE C1A 4P3, Canada
| | | | | |
Collapse
|
78
|
Lim J, Liu Z, Apontes P, Feng D, Pessin JE, Sauve AA, Angeletti RH, Chi Y. Dual mode action of mangiferin in mouse liver under high fat diet. PLoS One 2014; 9:e90137. [PMID: 24598864 PMCID: PMC3943915 DOI: 10.1371/journal.pone.0090137] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/28/2014] [Indexed: 12/31/2022] Open
Abstract
Chronic over-nutrition is a major contributor to the spread of obesity and its related metabolic disorders. Development of therapeutics has been slow compared to the speedy increase in occurrence of these metabolic disorders. We have identified a natural compound, mangiferin (MGF) (a predominant component of the plants of Anemarrhena asphodeloides and Mangifera indica), that can protect against high fat diet (HFD) induced obesity, hyperglycemia, insulin resistance and hyperlipidemia in mice. However, the molecular mechanisms whereby MGF exerts these beneficial effects are unknown. To understand MGF mechanisms of action, we performed unbiased quantitative proteomic analysis of protein profiles in liver of mice fed with HFD utilizing 15N metabolically labeled liver proteins as internal standards. We found that out of 865 quantified proteins 87 of them were significantly differentially regulated by MGF. Among those 87 proteins, 50% of them are involved in two major processes, energy metabolism and biosynthesis of metabolites. Further classification indicated that MGF increased proteins important for mitochondrial biogenesis and oxidative activity including oxoglutarate dehydrogenase E1 (Dhtkd1) and cytochrome c oxidase subunit 6B1 (Cox6b1). Conversely, MGF reduced proteins critical for lipogenesis such as fatty acid stearoyl-CoA desaturase 1 (Scd1) and acetyl-CoA carboxylase 1 (Acac1). These mass spectrometry data were confirmed and validated by western blot assays. Together, data indicate that MGF upregulates proteins pivotal for mitochondrial bioenergetics and downregulates proteins controlling de novo lipogenesis. This novel mode of dual pharmacodynamic actions enables MGF to enhance energy expenditure and inhibit lipogenesis, and thereby correct HFD induced liver steatosis and prevent adiposity. This provides a molecular basis supporting development of MGF or its metabolites into therapeutics to treat metabolic disorders.
Collapse
Affiliation(s)
- Jihyeon Lim
- The Laboratory for Macromolecular Analysis & Proteomics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- Department of Pathology, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- * E-mail: (JL)
| | - Zhongbo Liu
- Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- * E-mail: (JL)
| | - Pasha Apontes
- Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Daorong Feng
- Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Jeffrey E. Pessin
- Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Anthony A. Sauve
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, United States of America
| | - Ruth H. Angeletti
- The Laboratory for Macromolecular Analysis & Proteomics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- Department of Pathology, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Yuling Chi
- Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- * E-mail: (JL)
| |
Collapse
|
79
|
Zou B, Ge ZZ, Zhang Y, Du J, Xu Z, Li CM. Persimmon Tannin accounts for hypolipidemic effects of persimmon through activating of AMPK and suppressing NF-κB activation and inflammatory responses in High-Fat Diet Rats. Food Funct 2014; 5:1536-46. [DOI: 10.1039/c3fo60635j] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
High molecular weight persimmon tannin is a central component accounting for the anti-hyperlipidemic effects of consuming persimmon fruits via AMPK pathway.
Collapse
Affiliation(s)
- Bo Zou
- College of Food Science and Technology
- Huazhong Agricultural University
- Wuhan, China
| | - Zhen-zhen Ge
- College of Food Science and Technology
- Huazhong Agricultural University
- Wuhan, China
| | - Ying Zhang
- College of Food Science and Technology
- Huazhong Agricultural University
- Wuhan, China
| | - Jing Du
- College of Food Science and Technology
- Huazhong Agricultural University
- Wuhan, China
| | - Ze Xu
- College of Food Science and Technology
- Huazhong Agricultural University
- Wuhan, China
| | - Chun-mei Li
- College of Food Science and Technology
- Huazhong Agricultural University
- Wuhan, China
- Key Laboratory of Environment Correlative Food Science (Huazhong Agricultural University)
- Ministry of Education
| |
Collapse
|
80
|
Ma H, Chen H, Sun L, Tong L, Zhang T. Improving permeability and oral absorption of mangiferin by phospholipid complexation. Fitoterapia 2013; 93:54-61. [PMID: 24220727 DOI: 10.1016/j.fitote.2013.10.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 12/18/2022]
Abstract
Mangiferin is an active ingredient of medicinal plant with poor hydrophilicity and lipophilicity. Many reports focused on improving aqueous solubility, but oral bioavailability of mangiferin was still limited. In this study, we intended to increase not only solubility, but also membrane permeability of mangiferin by a phospholipid complexation technique. The new complex's physicochemical properties were characterized in terms of scanning electron microscopy (SEM), differential scanning calorimetry (DSC), infrared absorption spectroscopy (IR), aqueous solubility, oil-water partition coefficient and in vitro dissolution. The intestinal absorption of the complex was studied by the rat in situ intestinal perfusion model. After oral administration of mangiferin-phospholipid complex and crude mangiferin in rats, the concentrations of mangiferin were determined by a validated RP-HPLC method. Results showed that the solubility of the complex in water and in n-octanol was enhanced and the oil-water partition coefficient was improved by 6.2 times and the intestinal permeability in rats was enhanced significantly. Peak plasma concentration and AUC of mangiferin from the complex (Cmax: 377.66 μg/L, AUC: 1039.94 μg/L*h) were higher than crude mangiferin (Cmax: 180 μg/L, AUC: 2355.63 μg/L*h). In view of improved solubility and enhanced permeability, phospholipid complexation technique can increase bioavailability of mangiferin by 2.3 times in comparison to the crude mangiferin.
Collapse
Affiliation(s)
- Hequn Ma
- Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, China
| | - Hongming Chen
- Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, China
| | - Le Sun
- Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, China
| | - Lijin Tong
- Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, China
| | - Tianhong Zhang
- Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang, China.
| |
Collapse
|
81
|
Telang M, Dhulap S, Mandhare A, Hirwani R. Therapeutic and cosmetic applications of mangiferin: a patent review. Expert Opin Ther Pat 2013; 23:1561-80. [PMID: 24066838 DOI: 10.1517/13543776.2013.836182] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Mangiferin, a natural C-glucoside xanthone [2-C-β-D-glucopyranosyl-1, 3, 6, 7-tetrahydroxyxanthone], is abundantly present in young leaves and stem bark of the mango tree. The xanthonoid structure of mangiferin with C-glycosyl linkage and polyhydroxy components contributes to its free radical-scavenging ability, leading to a potent antioxidant effect as well as multiple biological activities. AREAS COVERED An extensive search was carried out to collect patent information on mangiferin and its derivatives using various patent databases spanning all priority years to date. The patents claiming therapeutic and cosmetic applications of mangiferin and its derivatives were analyzed in detail. The technology areas covered in this article include metabolic disorders, cosmeceuticals, multiple uses of the same compound, miscellaneous uses, infectious diseases, inflammation, cancer and autoimmune disorders, and neurological disorders. EXPERT OPINION Mangiferin has the potential to modulate multiple molecular targets including nuclear factor-kappa B (NF-κB) signaling and cyclooxygenase-2 (COX-2) protein expression. Mangiferin exhibits antioxidant, antidiabetic, antihyperuricemic, antiviral, anticancer and antiinflammatory activities. The molecular structure of mangiferin fulfils the four Lipinski's requisites reported to favor high bioavailability by oral administration. There is no evidence of adverse side effects of mangiferin so far. Mangiferin could thus be a promising candidate for development of a multipotent drug.
Collapse
Affiliation(s)
- Manasi Telang
- CSIR Unit for Research and Development of Information Products , "Jopasana", 85/1, Paud Road, Kothrud, Pune 411 038 , India +91 9371161742 ; +91 20 25383558 ;
| | | | | | | |
Collapse
|
82
|
Danthron activates AMP-activated protein kinase and regulates lipid and glucose metabolism in vitro. Acta Pharmacol Sin 2013; 34:1061-9. [PMID: 23770982 DOI: 10.1038/aps.2013.39] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/20/2013] [Indexed: 01/05/2023]
Abstract
AIM To discover the active compound on AMP-activated protein kinase (AMPK) activation and investigate the effects of the active compound 1,8-dihydroxyanthraquinone (danthron) from the traditional Chinese medicine rhubarb on AMPK-mediated lipid and glucose metabolism in vitro. METHODS HepG2 and C2C12 cells were used. Cell viability was determined using MTT assay. Real-time PCR was performed to measure the gene expression. Western blotting assay was applied to investigate the protein phosphorylation level. Enzymatic assay kits were used to detect the total cholesterol (TC), triglyceride (TG) and glucose contents. RESULTS Danthron (0.1, 1, and 10 μmol/L) dose-dependently promoted the phosphorylation of AMPK and acetyl-CoA carboxylase (ACC) in both HepG2 and C2C12 cells. Meanwhile, danthron treatment significantly reduced the lipid synthesis related sterol regulatory element-binding protein 1c (SREBP1c) and fatty acid synthetase (FAS) gene expressions, and the TC and TG levels. In addition, danthron treatment efficiently increased glucose consumption. The actions of danthron on lipid and glucose metabolism were abolished or reversed by co-treatment with the AMPK inhibitor compound C. CONCLUSION Danthron effectively reduces intracellular lipid contents and enhanced glucose consumption in vitro via activation of AMPK signaling pathway.
Collapse
|
83
|
Zhang Y, Liu X, Han L, Gao X, Liu E, Wang T. Regulation of lipid and glucose homeostasis by mango tree leaf extract is mediated by AMPK and PI3K/AKT signaling pathways. Food Chem 2013; 141:2896-905. [PMID: 23871039 DOI: 10.1016/j.foodchem.2013.05.121] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 04/14/2013] [Accepted: 05/24/2013] [Indexed: 11/16/2022]
Abstract
Ethanolic extract of Mangifera indica (mango) dose-dependently decreased serum glucose and triglyceride in KK-A(y) mice. Our in vitro and in vivo investigations revealed that the effect of mango leave extract (ME) on glucose and lipid homeostasis is mediated, at least in part, through the PI3K/AKT and AMPK signaling pathway. ME up-regulated the expression of PI3K, AKT and GYS genes by 2.0-fold, 3.2-fold, and 2.7-fold, respectively, leading to a decrease in glucose level. On the other hand, ME up-regulated AMPK and altered lipid metabolism. ME also down-regulated ACC (2.8-fold), HSL (1.6-fold), FAS (1.8-fold) and PPAR-γ (4.0-fold). Finally, we determined that active metabolites of benzophenone C-glucosides, Iriflophenone 3-C-β-glucoside and Foliamangiferoside A from ME, may play a dominant role in this integrated regulation of sugar and lipid homeostasis.
Collapse
Affiliation(s)
- Yi Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China
| | | | | | | | | | | |
Collapse
|
84
|
Lee KM, Yang SJ, Kim YD, Choi YD, Nam JH, Choi CS, Choi HS, Park CS. Disruption of the cereblon gene enhances hepatic AMPK activity and prevents high-fat diet-induced obesity and insulin resistance in mice. Diabetes 2013; 62:1855-64. [PMID: 23349485 PMCID: PMC3661653 DOI: 10.2337/db12-1030] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A nonsense mutation in cereblon (CRBN) causes a mild type of mental retardation in humans. An earlier study showed that CRBN negatively regulates the functional activity of AMP-activated protein kinase (AMPK) in vitro by binding directly to the α1-subunit of the AMPK complex. However, the in vivo role of CRBN was not studied. For elucidation of the physiological functions of Crbn, a mouse strain was generated in which the Crbn gene was deleted throughout the whole body. In Crbn-deficient mice fed a normal diet, AMPK in the liver showed hyperphosphorylation, which indicated the constitutive activation of AMPK. Since Crbn-deficient mice showed significantly less weight gain when fed a high-fat diet and their insulin sensitivity was considerably improved, the functions of Crbn in the liver were primarily investigated. These results provide the first in vivo evidence that Crbn is a negative modulator of AMPK, which suggests that Crbn may be a potential target for metabolic disorders of the liver.
Collapse
Affiliation(s)
- Kwang Min Lee
- School of Life Sciences and Cell Dynamics Research Center and National Leading Research Laboratory for Ion Channels, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Seung-Joo Yang
- School of Life Sciences and Cell Dynamics Research Center and National Leading Research Laboratory for Ion Channels, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yong Deuk Kim
- National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Yoo Duk Choi
- Department of Pathology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jong Hee Nam
- Department of Pathology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Incheon, Republic of Korea
- Division of Endocrinology, Gil Medical Center, Gachon University, Incheon, Republic of Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Chul-Seung Park
- School of Life Sciences and Cell Dynamics Research Center and National Leading Research Laboratory for Ion Channels, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- Corresponding author: Chul-Seung Park,
| |
Collapse
|
85
|
Zhang X, Wu C, Wu H, Sheng L, Su Y, Zhang X, Luan H, Sun G, Sun X, Tian Y, Ji Y, Guo P, Xu X. Anti-hyperlipidemic effects and potential mechanisms of action of the caffeoylquinic acid-rich Pandanus tectorius fruit extract in hamsters fed a high fat-diet. PLoS One 2013; 8:e61922. [PMID: 23613974 PMCID: PMC3628350 DOI: 10.1371/journal.pone.0061922] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 03/15/2013] [Indexed: 12/20/2022] Open
Abstract
Hyperlipidemia is considered to be one of the greatest risk factors contributing to the prevalence and severity of cardiovascular diseases. In this work, we investigated the anti-hyperlipidemic effect and potential mechanism of action of the Pandanus tectorius fruit extract in hamsters fed a high fat-diet (HFD). The n-butanol fraction of the P. tectorius fruit ethanol extract (PTF-b) was rich in caffeoylquinic acids (CQAs). Administration of PTF-b for 4 weeks effectively decreased retroperitoneal fat and the serum levels of total cholesterol (TC), triglycerides (TG) and low density lipoprotein-cholesterol (LDL-c) and hepatic TC and TG. The lipid signals (fatty acids, and cholesterol) in the liver as determined by nuclear magnetic resonance (NMR) were correspondingly reduced. Realtime quantitative PCR showed that the mRNA levels of PPARα and PPARα-regulated genes such as ACO, CPT1, LPL and HSL were largely enhanced by PTF-b. The transcription of LDLR, CYP7A1, and PPARγ was also upregulated. Treatment with PTF-b significantly stimulated the activation of AMP-activated protein kinase (AMPK) as well as the activity of serum and hepatic lipoprotein lipase (LPL). Together, these results suggest that administration of the PTF-b enriched in CQAs moderates hyperlipidemia and improves the liver lipid profile. These effects may be caused, at least in part, by increasing the expression of PPARα and its downstream genes and by upregulation of LPL and AMPK activities.
Collapse
Affiliation(s)
- Xiaopo Zhang
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chongming Wu
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Haifeng Wu
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | - Yan Su
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Research Centre on Life Sciences and Environment Sciences, Harbin University of Commerce, Harbin, China
| | - Xue Zhang
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Research Centre on Life Sciences and Environment Sciences, Harbin University of Commerce, Harbin, China
| | - Hong Luan
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Research Centre on Life Sciences and Environment Sciences, Harbin University of Commerce, Harbin, China
| | - Guibo Sun
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaobo Sun
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yu Tian
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yubin Ji
- Research Centre on Life Sciences and Environment Sciences, Harbin University of Commerce, Harbin, China
| | - Peng Guo
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- * E-mail: (PG); (XX)
| | - Xudong Xu
- Pharmacology and Toxicology Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- * E-mail: (PG); (XX)
| |
Collapse
|
86
|
Dudhia Z, Louw J, Muller C, Joubert E, de Beer D, Kinnear C, Pheiffer C. Cyclopia maculata and Cyclopia subternata (honeybush tea) inhibits adipogenesis in 3T3-L1 pre-adipocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:401-408. [PMID: 23428403 DOI: 10.1016/j.phymed.2012.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/01/2012] [Accepted: 12/15/2012] [Indexed: 06/01/2023]
Abstract
The stems, leaves and flowers of Cyclopia have been consumed as a herbal tea 'honeybush tea' to treat various medical ailments since the 19th century. Plant polyphenols are reported to inhibit adipogenesis in cell and animal models of obesity. The aim of this study was to assess the effect of hot water extracts of two Cyclopia species, C. maculata and C. subternata on obesity in an in vitro model. The total polyphenol content of unfermented C. subternata, unfermented C. maculata and fermented C. maculata extracts was 25.6, 22.4 and 10.8g GAE/100g, respectively. The major compounds present in the extracts were: the flavonoid, phloretin-3',5'-di-C-glucoside in C. subternata, the xanthone, mangiferin in unfermented C. maculata and the flavanone, hesperidin in fermented C. maculata. All of the plant extracts inhibited intracellular triglyceride and fat accumulation, and decreased PPARγ2 expression. The higher concentrations of unfermented C. maculata (800 and 1600μg/ml) and C. subternata (1600μg/ml) were cytotoxic in terms of decreased mitochondrial dehydrogenase activity. Both fermented and unfermented C. maculata, at concentrations greater than 100μg/ml, decreased cellular ATP content. Cyclopia maculata and C. subternata inhibit adipogenesis in vitro, suggesting their potential as anti-obesity agents.
Collapse
Affiliation(s)
- Zulfaqar Dudhia
- Diabetes Discovery Platform, Medical Research Council, Tygerberg, South Africa
| | | | | | | | | | | | | |
Collapse
|
87
|
Hou J, Zheng D, Fan K, Yu B, Xiao W, Ma J, Jin W, Tan Y, Wu J. Combination of Mangiferin and Dipeptidyl Peptidase-4 Inhibitor Sitagliptin Improves Impaired Glucose Tolerance in Streptozotocin-Diabetic Rats. Pharmacology 2012; 90:177-82. [DOI: 10.1159/000342128] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 07/17/2012] [Indexed: 01/07/2023]
|