1
|
Kim JT, Chen J, Zhou Y, Son MJ, Jeon DH, Kwon JW, Lee GY, Lee HJ. Cycloastragenol inhibits adipogenesis and fat accumulation in vitro and in vivo through activating Hedgehog signaling. Food Sci Biotechnol 2024; 33:711-720. [PMID: 38274180 PMCID: PMC10805729 DOI: 10.1007/s10068-023-01403-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 01/27/2024] Open
Abstract
In this study, we investigated the effect of cycloastragenol (CAG), a triterpenoid isolated from Astragalus membranaceus roots, on regulating the adipogenesis and fat accumulation in vitro and in vivo. During the adipogenesis of 3T3-L1 cells, CAG inhibited lipid accumulation and the expression of key adipogenic factors, proliferator-activated receptor γ (PPARγ) and CCAAT enhancer binding protein α (C/EBPα) and increased the expression of Gli1, a key mediator in Hedgehog (Hh) signaling. In HFD-induced animal experiment, CAG significantly reduced body weight gain without affecting brown fat weight. In addition, CAG regulated the expression of PPARγ, C/EBPα, and Gli1 in visceral white adipose tissue (vWAT). We also confirmed the inhibitory effect of CAG on specifically targeting white adipose tissue (WAT) formation in stromal vascular fraction (SVF) cell differentiation. Taken together, these results suggest that CAG may be a potent phytochemical preventing adipogenesis and obesity via Hh signaling. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01403-0.
Collapse
Affiliation(s)
- Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Jing Chen
- Institute for Advanced and Applied Chemical Synthesis, Jinan University, Guangzhou, 510632 China
| | - Yimeng Zhou
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Moon Jeong Son
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Dong Hyeon Jeon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Jung Won Kwon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Ga Yeon Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 456-756 South Korea
| |
Collapse
|
2
|
Li JZ, Chen N, Ma N, Li MR. Mechanism and Progress of Natural Products in the Treatment of NAFLD-Related Fibrosis. Molecules 2023; 28:7936. [PMID: 38067665 PMCID: PMC10707854 DOI: 10.3390/molecules28237936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most prevalent chronic liver disorder worldwide, with liver fibrosis (LF) serving as a pivotal juncture in NAFLD progression. Natural products have demonstrated substantial antifibrotic properties, ushering in novel avenues for NAFLD treatment. This study provides a comprehensive review of the potential of natural products as antifibrotic agents, including flavonoids, polyphenol compounds, and terpenoids, with specific emphasis on the role of Baicalin in NAFLD-associated fibrosis. Mechanistically, these natural products have exhibited the capacity to target a multitude of signaling pathways, including Hedgehog, Wnt/β-catenin, TGF-β1, and NF-κB. Moreover, they can augment the activities of antioxidant enzymes, inhibit pro-fibrotic factors, and diminish fibrosis markers. In conclusion, this review underscores the considerable potential of natural products in addressing NAFLD-related liver fibrosis through multifaceted mechanisms. Nonetheless, it underscores the imperative need for further clinical investigation to authenticate their effectiveness, offering invaluable insights for future therapeutic advancements in this domain.
Collapse
Affiliation(s)
- Jin-Zhong Li
- Division of Infectious Disease, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Ning Chen
- General Medicine, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Nan Ma
- Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Min-Ran Li
- Division of Infectious Disease, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| |
Collapse
|
3
|
Zheng P, Ding Y, Lu F, Liu N, Wu H, Bian Z, Chen X, Yang D. Atorvastatin reverses high cholesterol-induced cardiac remodelling and regulates mitochondrial quality-control in a cholesterol-independent manner: An experimental study. Clin Exp Pharmacol Physiol 2021; 48:1150-1161. [PMID: 33891707 DOI: 10.1111/1440-1681.13507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/09/2021] [Accepted: 04/17/2021] [Indexed: 01/03/2023]
Abstract
Mitochondria are key regulators of cell fate, maintaining self-stability by a fine-tuned quality-control network including mitophagy, biogenesis, fission and fusion processes. Myocardial mitochondria can be impaired by hypercholesterolemia. Statins, such as atorvastatin, are considered the cornerstone in the management of hypercholesterolaemia primarily due to their marked cholesterol-lowering ability. The direct effect of atorvastatin on myocardial mitochondria remains unclear. We aimed to explore whether atorvastatin could attenuate myocardial mitochondrial defects induced by high cholesterol, and whether cycloastragenol, a potent telomerase activator, could be used as a potential complementary bioactive compound for obesity and hypercholesterolaemia treatment. We found that atorvastatin at a low dose (3 mg/kg) did not reduce elevated serum cholesterol, but reversed cardiac remodelling and dysfunction in C57BL/6J mice fed with high-fat diet (HFD). Atorvastatin reversed the upregulated mitophagy, mitochondrial fission and fusion, accompanied by mitochondrial biogenesis activation in HFD-fed mice hearts. Mitochondrial structural impairments were attenuated by atorvastatin in HFD-fed mice and oxidized low-density lipoprotein (ox-LDL) exposed HL-1 cardiomyocytes. The depolarized mitochondrial membrane potential and increased mitochondrial oxygen consumption rates in ox-LDL exposed HL-1 cells were recovered by atorvastatin. Furthermore, atorvastatin co-treated with cycloastragenol had better effects on reducing body weight, improving cardiac remodelling and dysfunction, and protecting mitochondria in high cholesterol. Conclusively, low-dose atorvastatin exhibited a cholesterol-independent cardioprotective effect through improving the mitochondrial quality-control network and repairing mitochondrial ultrastructure in high cholesterol. Atorvastatin plus cycloastragenol supplement therapy has a better effect on treating obesity and hypercholesterolaemia.
Collapse
Affiliation(s)
- Peng Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanzi Ding
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feiyan Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nannan Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hengfang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiping Bian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangjian Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Science and Technology Office, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
J U C, Mohan MC, Prakash Kumar B. Attenuation of obesity related inflammation in RAW 264.7 macrophages and 3T3-L1 adipocytes by varanadi kashayam and identification of potential bioactive molecules by UHPLC-Q-Orbitrap HRMS. Arch Physiol Biochem 2021:1-15. [PMID: 33539199 DOI: 10.1080/13813455.2021.1877309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Obesity is associated with chronic low-grade inflammation, characterised by the up-regulation of pro-inflammatory cytokines in obese adipose tissue. In this study, 3T3-L1 CM enhanced TNF-α and IL-1β in RAW 264.7 cells compared with LPS treated cells. However, treatment with Varanadi kashayam suppressed the inflammatory changes associated with RAW 264.7 cells. Subsequently, RAW CM used to stimulate adipocytes, resulting in decreased intracellular lipid content and reduced adipogenic markers after Varanadi kashayam treatment. The chemical profiling of Varanadi kashayam using UHPLC-Q-Orbitrap-HRMS identified 194 compounds by comparing their retention time, the experimentally measured exact mass of precursor, and fragmented ions, and fragmentation pattern with spectral library and reported literature. Collectively, Varanadi kashayam act as a potent anti-inflammatory and anti-adipogenic agent that could disrupt the crosstalk between adipocytes and macrophages. Hence it could be a better candidate for reducing inflammation associated with obese adipose tissue.
Collapse
Affiliation(s)
- Chinchu J U
- Inflammation Research Lab, School of Biosciences, Mahatma Gandhi University, Kottayam, India
| | - Mohind C Mohan
- Inflammation Research Lab, School of Biosciences, Mahatma Gandhi University, Kottayam, India
| | - B Prakash Kumar
- Inflammation Research Lab, School of Biosciences, Mahatma Gandhi University, Kottayam, India
| |
Collapse
|
5
|
Guru A, Issac PK, Velayutham M, Saraswathi NT, Arshad A, Arockiaraj J. Molecular mechanism of down-regulating adipogenic transcription factors in 3T3-L1 adipocyte cells by bioactive anti-adipogenic compounds. Mol Biol Rep 2021; 48:743-761. [PMID: 33275195 DOI: 10.1007/s11033-020-06036-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022]
Abstract
Obesity is growing at an alarming rate, which is characterized by increased adipose tissue. It increases the probability of many health complications, such as diabetes, arthritis, cardiac disease, and cancer. In modern society, with a growing population of obese patients, several individuals have increased insulin resistance. Herbal medicines are known as the oldest method of health care treatment for obesity-related secondary health issues. Several traditional medicinal plants and their effective phytoconstituents have shown anti-diabetic and anti-adipogenic activity. Adipose tissue is a major site for lipid accumulation as well as the whole-body insulin sensitivity region. 3T3-L1 cell line model can achieve adipogenesis. Adipocyte characteristics features such as expression of adipocyte markers and aggregation of lipids are chemically induced in the 3T3-L1 fibroblast cell line. Differentiation of 3T3-L1 is an efficient and convenient way to obtain adipocyte like cells in experimental studies. Peroxisome proliferation activated receptor γ (PPARγ) and Cytosine-Cytosine-Adenosine-Adenosine-Thymidine/Enhancer-binding protein α (CCAAT/Enhancer-binding protein α or C/EBPα) are considered to be regulating adipogenesis at the early stage, while adiponectin and fatty acid synthase (FAS) is responsible for the mature adipocyte formation. Excess accumulation of these adipose tissues and lipids leads to obesity. Thus, investigating adipose tissue development and the underlying molecular mechanism is important in the therapeutical approach. This review describes the cellular mechanism of 3T3-L1 fibroblast cells on potential anti-adipogenic herbal bioactive compounds.
Collapse
Affiliation(s)
- Ajay Guru
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Praveen Kumar Issac
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Manikandan Velayutham
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - N T Saraswathi
- Molecular Biophysics Lab, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India
| | - Aziz Arshad
- International Institute of Aquaculture and Aquatic Sciences (I-AQUAS), Universiti Putra Malaysia, 71050, Port Dickson, Negeri Sembilan, Malaysia
- Department of Aquaculture, Faculty of Agriculture, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Jesu Arockiaraj
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
6
|
Proshkina E, Plyusnin S, Babak T, Lashmanova E, Maganova F, Koval L, Platonova E, Shaposhnikov M, Moskalev A. Terpenoids as Potential Geroprotectors. Antioxidants (Basel) 2020; 9:antiox9060529. [PMID: 32560451 PMCID: PMC7346221 DOI: 10.3390/antiox9060529] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/09/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023] Open
Abstract
Terpenes and terpenoids are the largest groups of plant secondary metabolites. However, unlike polyphenols, they are rarely associated with geroprotective properties. Here we evaluated the conformity of the biological effects of terpenoids with the criteria of geroprotectors, including primary criteria (lifespan-extending effects in model organisms, improvement of aging biomarkers, low toxicity, minimal adverse effects, improvement of the quality of life) and secondary criteria (evolutionarily conserved mechanisms of action, reproducibility of the effects on different models, prevention of age-associated diseases, increasing of stress-resistance). The number of substances that demonstrate the greatest compliance with both primary and secondary criteria of geroprotectors were found among different classes of terpenoids. Thus, terpenoids are an underestimated source of potential geroprotectors that can effectively influence the mechanisms of aging and age-related diseases.
Collapse
Affiliation(s)
- Ekaterina Proshkina
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
| | - Sergey Plyusnin
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky Prosp., 167001 Syktyvkar, Russia
| | - Tatyana Babak
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
| | - Ekaterina Lashmanova
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
| | | | - Liubov Koval
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky Prosp., 167001 Syktyvkar, Russia
| | - Elena Platonova
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky Prosp., 167001 Syktyvkar, Russia
| | - Mikhail Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Centre, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russia; (E.P.); (S.P.); (T.B.); (E.L.); (L.K.); (E.P.); (M.S.)
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky Prosp., 167001 Syktyvkar, Russia
- Correspondence: ; Tel.: +7-8212-312-894
| |
Collapse
|
7
|
Yu Y, Wu J, Li J, Liu Y, Zheng X, Du M, Zhou L, Yang Y, Luo S, Hu W, Li L, Yao W, Liu Y. Cycloastragenol prevents age-related bone loss: Evidence in d-galactose-treated and aged rats. Biomed Pharmacother 2020; 128:110304. [PMID: 32497865 DOI: 10.1016/j.biopha.2020.110304] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/10/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIMS Aging-induced bone loss is a multifactorial, age-related, and progressive phenomenon among the general population and may further progress to osteoporosis and increase the risk of fractures. Cycloastragenol (CAG), currently the only compound reported that activates human telomerase, is thought to be able to alleviate or delay the symptoms of aging and chronic diseases. Previous research has suggested that CAG may have the potential to alleviate age-related bone loss. However, to date, no research has specifically focused on this aspect. In this study, we aimed to investigate whether CAG could prevent senile osteoporosis, and further reveal its underlying mechanism. METHODS CAG treatment was administrated into two bone loss rat models (D-galactose administration and aging) for 20 weeks and 33 weeks, respectively. Serum biomarkers analyses, bone biomechanical tests, micro-computed tomography assessment, and bone histomorphometry analyses were performed on the bone samples collected at the endpoint, to determine whether CAG could prevent or alleviate age-related bone loss. Proteomic analysis was performed to reveal the changes in protein profiles of the bones, and western blot was used to further verify the identity of the key proteins. The viability, osteoblastic differentiation, and mineralization of MC3T3-E1 cells were also evaluated after CAG treatment in vitro. RESULTS The results suggest that CAG treatment improves bone formation, reduces osteoclast number, alleviates the degradation of bone microstructure, and enhances bone biomechanical properties in both d-galactose- and aging-induced bone loss models. CAG treatment promotes viability, osteoblastic differentiation, and mineralization in MC3T3-E1 cells. Proteomic and western blot analyses revealed that CAG treatment increases osteoactivin (OA) expression to alleviate bone loss. CONCLUSION The results revealed that CAG alleviates age-related bone loss and improves bone microstructure and biomechanical properties. This may due to CAG-induced increase in OA expression. In addition, the results support preclinical investigations of CAG as a potential therapeutic medicine for the treatment of senile osteoporosis.
Collapse
Affiliation(s)
- Yongjie Yu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Jingkai Wu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Jin Li
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Yanzhi Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Medical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China; Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, PR China
| | - Xiaoyan Zheng
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Mingzhu Du
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Limin Zhou
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Shiying Luo
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Wenjia Hu
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China
| | - Lin Li
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Weimin Yao
- Department of Respiratory Medicine, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, PR China.
| | - Yuyu Liu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong, 524023, PR China.
| |
Collapse
|
8
|
Exploring Molecular Mechanism of Huangqi in Treating Heart Failure Using Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6473745. [PMID: 32382301 PMCID: PMC7195658 DOI: 10.1155/2020/6473745] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/05/2019] [Accepted: 01/06/2020] [Indexed: 11/17/2022]
Abstract
Heart failure (HF), a clinical syndrome with a high incidence due to various reasons, is the advanced stage of most cardiovascular diseases. Huangqi is an effective treatment for cardiovascular disease, which has multitarget, multipathway functions. Therefore, we used network pharmacology to explore the molecular mechanism of Huangqi in treating HF. In this study, 21 compounds of Huangqi, which involved 407 targets, were obtained and reconfirmed using TCMSP and PubChem databases. Moreover, we used Cytoscape 3.7.1 to construct compound-target network and screened the top 10 compounds. 378 targets related to HF were obtained from CTD and GeneCards databases and HF-target network was constructed by Cytoscape 3.7.1. The 46 overlapping targets of HF and Huangqi were gotten by Draw Venn Diagram. STRING database was used to set up a protein-protein interaction network, and MCODE module and the top 5 targets with the highest degree for overlapping targets were obtained. GO analysis performed by Metascape indicated that the overlapping targets were mainly enriched in blood vessel development, reactive oxygen species metabolic process, response to wounding, blood circulation, and so on. KEGG analysis analyzed by ClueGO revealed that overlapping targets were mainly enriched in AGE-RAGE signaling pathway in diabetic complications, IL-17 signaling pathway, HIF-1 signaling pathway, c-type lectin receptor signaling pathway, relaxin signaling pathway, and so on. Finally, molecular docking showed that top 10 compounds of Huangqi also had good binding activities to important targets compared with digoxin, which was carried out in CB-Dock molecular docking server. In conclusion, Huangqi has potential effect on regulating overlapping targets and GE-RAGE signaling pathway in diabetic complications, IL-17 signaling pathway, HIF-1 signaling pathway, and so on to be a latent multitarget, multipathway treatment for HF.
Collapse
|
9
|
J.U C, Mohan MC, B PK. Downregulation of adipogenic genes in 3T3-L1 Pre adipocytes- a possible mechanism of anti-obesity activity of herbal decoction Varanadi Kashayam. J Herb Med 2020. [DOI: 10.1016/j.hermed.2019.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Liu J, Gao D, Dan J, Liu D, Peng L, Zhou R, Luo Y. The protective effect of cycloastragenol on aging mouse circadian rhythmic disorder induced by d-galactose. J Cell Biochem 2019; 120:16408-16415. [PMID: 31310357 DOI: 10.1002/jcb.28587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/19/2019] [Accepted: 01/24/2019] [Indexed: 01/04/2023]
Abstract
Aging process in mammals is associated with a decline in amplitude and a long period of circadian behaviors which are regulated by a central circadian regulator in the suprachiasmatic nucleus (SCN) and local oscillators in peripheral tissues. It is unclear whether enhancing clock function can retard aging. Using fibroblasts expressing per2::lucSV and senescent cells, we revealed cycloastragenol (CAG), a natural aglycone derivative from astragaloside IV, as a clock amplitude enhancing small molecule. CAG could activate telomerase to antiaging, but no reports focused on its effects on circadian rhythm disorders in aging mice. Here we analyze the potential effects of CAG on d-galactose-induced aging mice on the circadian behavior and expression of clock genes. For this purpose, CAG (20 mg/kg orally), was administered daily to d-galactose (150 mg/kg, subcutaneous) mice model of aging for 6 weeks. An actogram analysis of free-running activity of these mice showed that CAG significantly enhances the locomotor activity. We further found that CAG increase expressions of per2 and bmal1 genes in liver and kidney of aging mouse. Furthermore, CAG enhanced clock protein BMAL1 and PER2 levels in aging mouse liver and SCN. Our results indicated that the CAG could restore the behavior of circadian rhythm in aging mice induced by d-galactose. These data of present study suggested that CAG could be used as a novel therapeutic strategy for the treatment of age-related circadian rhythm disruption.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Dongxiao Gao
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Dan Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Lei Peng
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Ruoyu Zhou
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Ying Luo
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| |
Collapse
|
11
|
Yu Y, Zhou L, Yang Y, Liu Y. Cycloastragenol: An exciting novel candidate for age-associated diseases. Exp Ther Med 2018; 16:2175-2182. [PMID: 30186456 PMCID: PMC6122403 DOI: 10.3892/etm.2018.6501] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022] Open
Abstract
Cycloastragenol (CAG) is a triterpenoid saponin compound and a hydrolysis product of the main active ingredient in Astragalus membranaceus (Fisch.) Bunge. An increasing body of evidence has indicated that CAG has a wide spectrum of pharmacological functions, which are attracting attention in the research community. The aim of the present review paper was to review and elucidate the advanced study of CAG. The focus was on advanced studies of CAG in English and Chinese databases; the literature was collected and reviewed to summarize the latest efficacy, pharmacokinetics and adverse reactions of CAG. Extensive pharmacological effects have been attributed to CAG, including telomerase activation, telomere elongation, anti-inflammatory and anti-oxidative properties; CAG has also been reported to improve lipid metabolism. Clinical research has demonstrated that CAG activates telomerase in humans and ameliorates various biomarkers. CAG is absorbed through the intestinal epithelium via passive diffusion and undergoes first-pass hepatic metabolism. Within a certain dose range, oral CAG is relatively safe; however, underlying mechanisms associated with CAG are not clear, and thus, we should be aware of potential adverse reactions associated with CAG. According to existing studies and clinical trials, CAG is safe and has broad application prospects. However, further studies are required to fully understand its efficacy and potential adverse reactions, and to ensure the proper use of CAG is applied to treat diseases clinically.
Collapse
Affiliation(s)
- Yongjie Yu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Limin Zhou
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yuyu Liu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| |
Collapse
|
12
|
Gu M, Zhang S, Zhao Y, Huang J, Wang Y, Li Y, Fan S, Yang L, Ji G, Tong Q, Huang C. Cycloastragenol improves hepatic steatosis by activating farnesoid X receptor signalling. Pharmacol Res 2017; 121:22-32. [PMID: 28428116 DOI: 10.1016/j.phrs.2017.04.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/16/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a global health problem. However, there is no approved therapy for NAFLD. Farnesoid X receptor (FXR) is a potential drug target for treatment of NAFLD. In an attempt to screen FXR agonists, we found that cycloastragenol (CAG), a natural occurring compound in Astragali Radix, stimulated FXR transcription activity. In animal studies, we demonstrated that CAG treatment resulted in obvious reduction of high-fat diet induced lipid accumulation in liver accompanied by lowered blood glucose, serum triglyceride levels and hepatic bile acid pool size. The stimulation of FXR signalling by CAG treatment in DIO mice was confirmed via gene expression and western blot analysis. Molecular docking data further supported the interaction of CAG and FXR. In addition, CAG alleviated hepatic steatosis in methionine and choline deficient L-amino acid diet (MCD) induced non-alcoholic steatohepatitis (NASH) mice. Our data suggest that CAG ameliorates NAFLD via the enhancement of FXR signalling.
Collapse
Affiliation(s)
- Ming Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiying Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Zhao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinwen Huang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, China
| | - Yahui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yin Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengjie Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological Sciences, University of Texas McGovern Medical School, Houston, TX, USA
| | - Li Yang
- Research Center for Traditional Chinese Medicine of Complexity Systems, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological Sciences, University of Texas McGovern Medical School, Houston, TX, USA.
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
13
|
Duan X, Meng Q, Wang C, Liu Z, Liu Q, Sun H, Sun P, Yang X, Huo X, Peng J, Liu K. Calycosin attenuates triglyceride accumulation and hepatic fibrosis in murine model of non-alcoholic steatohepatitis via activating farnesoid X receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 25:83-92. [PMID: 28190475 DOI: 10.1016/j.phymed.2016.12.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/28/2016] [Accepted: 12/12/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) represents the more severe end of hepatic steatosis and is associated with progressive liver disease. Calycosin, derived from the root of Radix Astragali, has been demonstrated to have favorable efficacy on acute liver injury. PURPOSE The present study was to investigate the hepatoprotective effect of calycosin on attenuating triglyceride accumulation and hepatic fibrosis, as well as explore the potential mechanism in murine model of NASH. STUDY DESIGN The C57BL/6 male mice were fed with methionine choline deficient (MCD) diet for 4 weeks to induce NASH and treated with or without calycosin by oral gavage for 4 weeks. METHODS The body weight, liver weight and the liver to body weight ratios were measured. Serum ALT, AST, TG, TC, FFA, MCP-1 and mKC levels were accessed by biochemical methods. H&E staining and Oil red O staining were used to identify the amelioration of liver histopathology. Immunohistochemistry of a-SMA, Masson trichrome staining and Sirius red staining were used to identify the amelioration of hepatic fibrosis. The quantitative real-time-PCR and Western blot were applied to observe the expression changes of key factors involved in triglyceride synthesis, free fatty acid β-oxidation and hepatic fibrosis. RESULTS Calycosin significantly inhibited body weight loss induced by MCD diet, decreased the ALT and AST activities, MCP-1 and mKC in a dose-dependent manner. The H&E and Oil red O staining indicated calycosin effectively improved hepatic steatosis, improved the degree of triglyceride accumulation. Masson trichrome and Sirius red staining indicated that calycosin treatment remarkably attenuated the degree of hepatic fibrosis. Immunohistochemistry of a-SMA demonstrated that calycosin attenuated hepatic fibrosis by inhibiting hepatic stellate cell activation. Further, calycosin inhibited the expression of SREBP-1c, FASN, ACC and SCD1 involved in triglyceride synthesis, promoted the expression of PPARa, CPT1, Syndecan-1 and LPL involved in free fatty acid β-oxidation. The above effects of calycosin were attributed to FXR activation. CONCLUSION Calycosin attenuates triglyceride accumulation and hepatic fibrosis to protect against NASH via FXR activation.
Collapse
Affiliation(s)
- Xingping Duan
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China.
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Zhihao Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Qi Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Pengyuan Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China
| | - Xiaobo Yang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Xiaokui Huo
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China
| | - Jinyong Peng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Lvshunkou District, Dalian, Liaoning, China; Provincial Key Laboratory for Pharmacokinetics and Transport, Dalian, Liaoning, China.
| |
Collapse
|
14
|
Pérez-Jiménez A, Rufino-Palomares EE, Fernández-Gallego N, Ortuño-Costela MC, Reyes-Zurita FJ, Peragón J, García-Salguero L, Mokhtari K, Medina PP, Lupiáñez JA. Target molecules in 3T3-L1 adipocytes differentiation are regulated by maslinic acid, a natural triterpene from Olea europaea. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1301-1311. [PMID: 27765349 DOI: 10.1016/j.phymed.2016.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/15/2016] [Accepted: 07/01/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Metabolic syndrome is a set of pathologies among which stand out the obesity, which is related to the lipid droplet accumulation and changes to cellular morphology regulated by several molecules and transcription factors. Maslinic acid (MA) is a natural product with demonstrated pharmacological functions including anti-inflammation, anti-tumor and anti-oxidation, among others. PURPOSE Here we report the effects of MA on the adipogenesis process in 3T3-L1 cells. METHODS Cell viability, glucose uptake, cytoplasmic triglyceride droplets, triglycerides quantification, gene transcription factors such as peroxisome proliferator-activated receptor γ (PPARγ) and adipocyte fatty acid-binding protein (aP2) and intracellular Ca2+ levels were determined in pre-adipocytes and adipocytes of 3T3-L1 cells. RESULTS MA increased glucose uptake. MA also decreased lipid droplets and triglyceride levels, which is in concordance with the down-regulation of PPARγ and aP2. Finally, MA increased the intracellular Ca2+ concentration, which could also be involved in the demonstrated antiadipogenic effect of this triterpene. CONCLUSION MA has been demonstrated as potential antiadipogenic compound in 3T3-L1 cells.
Collapse
Affiliation(s)
- Amalia Pérez-Jiménez
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain; Department of i+D+I, Biomaslinic S.L., Polígono Industrial de Escúzar, 18130 Granada, Spain.
| | - Eva E Rufino-Palomares
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain; Pfizer Pharmaceutical-University of Granada-Government of Andalusian, Centre of Genomic and Oncologic Investigation (GENyO), Technological Park of Health Sciences, 18016 Granada, Spain.
| | - Nieves Fernández-Gallego
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - M Carmen Ortuño-Costela
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Fernando J Reyes-Zurita
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Juan Peragón
- Department of Experimental Biology, Biochemistry Section, Faculty of Experimental Biology, University of Jaén, 23071 Jaén, Spain
| | - Leticia García-Salguero
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Khalida Mokhtari
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain; Department of Biology, Faculty of Sciences, Mohammed I University, BP 717 60000 Oujda, Morocco
| | - Pedro P Medina
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain; Pfizer Pharmaceutical-University of Granada-Government of Andalusian, Centre of Genomic and Oncologic Investigation (GENyO), Technological Park of Health Sciences, 18016 Granada, Spain
| | - José A Lupiáñez
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain.
| |
Collapse
|
15
|
Ma PK, Wei BH, Cao YL, Miao Q, Chen N, Guo CE, Chen HY, Zhang YJ. Pharmacokinetics, metabolism, and excretion of cycloastragenol, a potent telomerase activator in rats. Xenobiotica 2016; 47:526-537. [DOI: 10.1080/00498254.2016.1204568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Peng-Kai Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
| | - Bao-Hong Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
- National Engineering Research Center for Marine Drugs, Ocean University of China, Qingdao, China, and
| | - Yan-Ling Cao
- Research Center for Life Science and Environmental Sciences, Haerbin University of Commerce, Haerbin, China
| | - Qing Miao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
| | - Ning Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
| | - Chang-E Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
| | - Hong-Ying Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
| | - Yu-Jie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China,
| |
Collapse
|
16
|
Wang S, Zhang Q, Zhang Y, Shen C, Wang Z, Wu Q, Zhang Y, Li S, Qiao Y. Agrimol B suppresses adipogenesis through modulation of SIRT1-PPAR gamma signal pathway. Biochem Biophys Res Commun 2016; 477:454-60. [PMID: 27320865 DOI: 10.1016/j.bbrc.2016.06.078] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 06/15/2016] [Indexed: 11/17/2022]
Abstract
Studies of human genetics have implicated the role of SIRT1 in regulating obesity, insulin resistance, and longevity. These researches motivated the identification of novel SIRT1 activators. The current study aimed to investigate the potential efficacy of agrimol B, a polyphenol derived from Agrimonia pilosa Ledeb., on mediating SIRT1 activity and fat metabolism. Results showed that agrimol B significantly induced cytoplasm-to-nucleus shuttle of SIRT1. Furthermore, we confirmed that agrimol B dramatically inhibited 3T3-L1 adipocyte differentiation by reducing PPARγ, C/EBPα, FAS, UCP-1, and apoE expression. Consequently, adipogenesis was blocked by treatment of agrimol B at the early stage of differentiation in a dose-dependent manner, the IC50 value was determined as 3.35 ± 0.32 μM. Taken together, our data suggest a therapeutic potential of agrimol B on alleviating obesity, through modulation of SIRT1-PPARγ signal pathway.
Collapse
Affiliation(s)
- Shifeng Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Qiao Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Yuxin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Cheng Shen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Zhen Wang
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qinghua Wu
- HD Biosciences Co., Ltd, Shanghai 201201, China
| | - Yanling Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Shiyou Li
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yanjiang Qiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
17
|
Identification of berbamine dihydrochloride from barberry as an anti-adipogenic agent by high-content imaging assay. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2016. [DOI: 10.1016/j.jtcms.2016.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
18
|
Nam MH, Lee SW, Na HY, Yoo JH, Paik SH, Ahn KS, Ahn YM, Ahn SY, Choi SH, Lee BC. Herbal Acupuncture for the Treatment of Obesity. J Acupunct Meridian Stud 2015; 9:49-57. [PMID: 27079225 DOI: 10.1016/j.jams.2015.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 12/29/2022] Open
Abstract
Obesity is the state of excessive body fat accumulation and is mainly caused by consuming more calories than are burned through physical activity. Herbal acupuncture (HA), also known as pharmacopuncture, has been increasingly used in clinics of Korean medical to alleviate obesity. This review analyzed four clinical studies and 16 animal studies on the effectiveness of HA as a treatment for obesity. Clinical evidence suggests that various kinds of HA might be beneficial for treating obesity; however, further investigations with well-designed, evidence-based, randomized clinical trials are needed. Animal studies support the idea that HA might be beneficial for the treatment of obesity and provide possible mechanisms, such as anti-inflammation, antioxidation, modulating lipid metabolism and so on, to explain the effect of HA on obesity. This review, based on the evidence collected, suggests that HA could have a beneficial effect for alleviating obesity by modulating inflammation, oxidative stress, lipid metabolism, leptin, and the insulin signal.
Collapse
Affiliation(s)
- Min-Ho Nam
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Seung-Wook Lee
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyun-Young Na
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jeong-Hwa Yoo
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Sun-Ho Paik
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Kwang Seok Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Young-Min Ahn
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Se-Young Ahn
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Seung-Hoon Choi
- Department of Medicine Consilience, Dankook University, Yongin, South Korea.
| | - Byung-Cheol Lee
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
19
|
Jin Y, Guo X, Yuan B, Yu W, Suo H, Li Z, Xu H. Disposition of Astragaloside IV via Enterohepatic Circulation Is Affected by the Activity of the Intestinal Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:6084-6093. [PMID: 26066785 DOI: 10.1021/acs.jafc.5b00168] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Astragaloside IV (ASIV) is a typical bioactive constituent of Radix Astragali. The study aimed to investigate the enterohepatic circulation of ASIV and evaluate the impact of activity of intestinal microbiota on the deposition of ASIV. The amounts of ASIV and its metabolites were quantified by an LC-MS/MS method. ASIV was metabolized by intestinal bacteria to form brachyoside B (Bra B), cyclogaleginoside B (Cyc B), cycloastragenol (CA), iso-cycloastragenol (iso-CA), and dehydrogenated metabolite of CA (CA-2H). CA and iso-CA circulated in blood besides ASIV when rats received ASIV intragastrically or intravenously. After rats were intragastrically administered 10 mg/kg ASIV, the AUC0-t values of ASIV, CA, and iso-CA were 109 ± 55, 26.8 ± 17.9, and 77.9 ± 35.1 nM·h, respectively. The plasma distribution of ASIV was significantly affected by bile duct drainage when ASIV was administered through the duodenum. ASIV, Bra B, and Cyc B were secreted from bile after duodenal administration of ASIV. Antibiotics markedly inhibited the metabolism of ASIV in intestinal microbiota. After rats were pretreated with antibiotics, the AUC0-t of iso-CA was 4.8 times less than that in control rats and the concentration of CA became undetectable. Variations in intestinal microbiota may change the disposition of ASIV and subsequently influence its potential health benefits.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xingjie Guo
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bo Yuan
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wenhong Yu
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hao Suo
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhiyuan Li
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haiyan Xu
- Department of Pharmaceutical Analysis, Pharmacy School, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
20
|
Gong AGW, Li N, Lau KM, Lee PSC, Yan L, Xu ML, Lam CTW, Kong AYY, Lin HQ, Dong TTX, Tsim KWK. Calycosin orchestrates the functions of Danggui Buxue Tang, a Chinese herbal decoction composing of Astragali Radix and Angelica Sinensis Radix: An evaluation by using calycosin-knock out herbal extract. JOURNAL OF ETHNOPHARMACOLOGY 2015; 168:150-157. [PMID: 25796405 DOI: 10.1016/j.jep.2015.03.033] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/07/2015] [Accepted: 03/10/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danggui Buxue Tang (DBT) is a classical Chinese herbal decoction containing two herbs, Astragali Radix (AR) and Angelicae Sinensis Radix (ASR), which serves as dietary supplement for treating women menopausal syndromes. Pharmacological studies indicate that DBT has estrogenic, erythropoietic and osteogenic properties; however, the action mechanism for this complex herbal decoction is not known. Calycosin, a major flavonoid in AR, shares similar structure with β-estradiol, and thus which is hypothesized to be the critical compound of DBT. Here, we aim to investigate the role of calycosin in DBT in terms of its biological functions by using a calycosin-depleted DBT decoction (DBT(Δcal)). The biological functions of DBT(Δcal) and parental DBT were systematically compared. MATERIALS AND METHODS In order to standardize DBT decoction, four chemical markers were determined and quantified by HPLC. A semi-preparative HPLC method was utilized to prepare DBT(Δcal). The authenticity of DBT(Δcal) was evaluated by LC-QQQ-MS/MS. To reveal the effect of calycosin on DBT functions, several cell assays related to the known properties of DBT were revealed, including estrogenic, erythropoietic and osteogenic functions. RESULTS As compared to parental DBT, the estrogenic, erythropoietic and osteogenic abilities were markedly reduced in DBT(Δcal). However, calycosin alone did not show significant responses. CONCLUSIONS Our results suggest that calycosin is a bioactive chemical in DBT decoction, and which could play a key linker in orchestrating multi-components of DBT as to achieve maximal functions. These discoveries should be invaluable in drug development and in investigating the modernization of traditional Chinese medicine from a new perspective.
Collapse
Affiliation(s)
- Amy Guo-wei Gong
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Ning Li
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Kei-man Lau
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Pinky Sum-chi Lee
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Lu Yan
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Miranda Li Xu
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Candy Ting-wai Lam
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Ava Yeung-yeung Kong
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Huang-quan Lin
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Tina Ting-xia Dong
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China
| | - Karl Wah-keung Tsim
- Division of Life Science, Center for Chinese Medicine R&D, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong Special Administrative Region, China.
| |
Collapse
|