1
|
Hu C, Nong S, Ke Q, Wu Z, Jiang Y, Wang Y, Chen Y, Wu Z, Zhang Q, Liao C, Wu M. Simultaneous co-delivery of Ginsenoside Rg3 and imiquimod from PLGA nanoparticles for effective breast cancer immunotherapy. iScience 2025; 28:112274. [PMID: 40256328 PMCID: PMC12008673 DOI: 10.1016/j.isci.2025.112274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/11/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Breast cancer is a fatal malignancy facing human health, with most patients experiencing recurrence and resistance to chemotherapy. The immunosuppressive tumor microenvironment (TME) greatly limits the actual outcome of immunotherapy. This study aimed to develop a modality of theranostics nanoparticles for breast cancer based on a near-infrared light-triggered nanoparticle for the targeted delivery of ginsenoside Rg3 and immune adjuvants imiquimod (R837) for effective breast cancer immunotherapy. Folate-receptor (FA) targeting IR780-R837/ginsenoside Rg3-perfluorohexane (PFH) @ polyethylene glycol (PEG)-poly (lactide-co-glycolic acid) (PLGA) nanoparticles (FA-NPs) can be activated by near-infrared laser irradiation in tumors, which leads to rapid release of ginsenoside Rg3 and R837 in the regions with high expression of folate receptors and glucose transporter 1 (GLUT1). Meanwhile, the nanoparticles can be used as dual-mode contrast agents for photoacoustic and ultrasound imaging. This strategy provides a strong immune memory effect, which can prevent tumor recurrence after eliminating the initial tumor.
Collapse
Affiliation(s)
- Cong Hu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Shuxiong Nong
- Department of Cardiology, Baise People’s Hospital. Affiliated Southwest Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Qianqian Ke
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ziming Wu
- School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuancheng Jiang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ying Wang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Yixin Chen
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Ziling Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Qi Zhang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| | - Chilin Liao
- Department of Cardiology, Baise People’s Hospital. Affiliated Southwest Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
2
|
Muhammad SNH, Ramli RR, Nik Mohamed Kamal NNS, Fauzi AN. Terpenoids: Unlocking Their Potential on Cancer Glucose Metabolism. Phytother Res 2024; 38:5626-5640. [PMID: 39300823 DOI: 10.1002/ptr.8346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
Cancer incidence has increased globally and has become the leading cause of death in the majority of countries. Many cancers have altered energy metabolism pathways, such as increased glucose uptake and glycolysis, as well as decreased oxidative phosphorylation. This is known as the Warburg effect, where cancer cells become more reliant on glucose to generate energy and produce lactate as an end product, even when oxygen is present. These are attributed to the overexpression of key glycolytic enzymes, glucose transporters, and related signaling pathways that occur in cancer cells. Therefore, overcoming metabolic alterations in cancer cells has recently become a target for therapeutic approaches. Natural products have played a key role in drug discovery, especially for cancer and infectious diseases. In this review, we are going to focus on terpenoids, which are gradually gaining popularity among drug researchers due to their reported anti-cancer effects via cell cycle arrest, induction of apoptosis, reduction of proliferation, and metastasis. This review summarizes the potential of 13 terpenoid compounds as anti-glycolytic inhibitors in different cancer models, primarily by inhibiting the glucose uptake and the generation of lactate, as well as by downregulating enzymes associated to glycolysis. As a conclusion, disruption of cancer cell glycolysis may be responsible for the anti-cancer activity of terpenoids.
Collapse
Affiliation(s)
- Siti Nur Hasyila Muhammad
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Redzyque Ramza Ramli
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Nik Nur Syazni Nik Mohamed Kamal
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Pulau Pinang, Malaysia
| | - Agustine Nengsih Fauzi
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
3
|
Guo C, Su Y, Wang H, Cao M, Diao N, Liu Z, Chen D, Kong M. A novel saponin liposomes based on the couplet medicines of Platycodon grandiflorum-Glycyrrhiza uralensis for targeting lung cancer. Drug Deliv 2022; 29:2743-2750. [PMID: 35999702 PMCID: PMC9487977 DOI: 10.1080/10717544.2022.2112997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Liposomes have been widely used for targeted drug delivery, but the disadvantages caused by cholesterol limit the application of conventional liposomes in cancer treatment. The compatibility basis of couplet medicines and the compatibility principle of the traditional Chinese medicine principle of ‘monarch, minister, assistant and guide’ are the important theoretical basis of Chinese medicine in the treatment of tumor and the important method to solve the problem of high toxicity. In this study, the active ingredients of the couplet medicines Platycodon grandiflorum and Glycyrrhiza uralensis were innovatively utilized, and glycyrrhizic acid (GA) was encapsulated in liposomes constructed by mixing saponin and lecithin, and cholesterol was replaced by platycodin and ginsenoside to construct saponin liposomes (RP-lipo) for the drug delivery system of Chinese medicine. Compared with conventional liposomes, PR-lipo@GA has no significant difference in morphological characteristics and drug release behavior, and also shows stronger targeting of lung cancer cells and anti-tumor ability in vitro, which may be related to the pharmacological properties of saponins themselves. Thus, PR-lipo@GA not only innovatively challenges the status of cholesterol as a liposome component, but also provides another innovative potential system with multiple functions for the clinical application of TCM couplet medicines.
Collapse
Affiliation(s)
- Chunjing Guo
- College of Marine Life Science, Ocean University of China, Qingdao266003, P.R. China
| | - Yanguo Su
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Hui Wang
- Gynecology Department, Affiliated hospital of Weifang Medical University, Weifang261053, P.R. China
| | - Min Cao
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Ningning Diao
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Zhongxin Liu
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Daquan Chen
- School of Pharmacy, Yantai University, Yantai264005, P.R. China
| | - Ming Kong
- College of Marine Life Science, Ocean University of China, Qingdao266003, P.R. China
| |
Collapse
|
4
|
Gao F, Fu Y, Yi J, Gao A, Jia Y, Cai S. Effects of Different Dietary Flavonoids on Dipeptidyl Peptidase-IV Activity and Expression: Insights into Structure-Activity Relationship. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12141-12151. [PMID: 33063510 DOI: 10.1021/acs.jafc.0c04974] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The inhibitory effects of 30 dietary flavonoids on dipeptidyl peptidase-IV (DPP-IV) were investigated to illustrate their quantitative structure-activity relationship (QSAR) and further explore their inhibition at the cellular level. Results of in vitro experiment show that isorhamnetin-3-O-glucoside (IC50, 6.53 ± 0.280 μM) had the strongest inhibition followed by cyanidin-3-O-glucoside (IC50, 8.26 ± 0.143 μM) and isorhamnetin-3-O-rutinoside (IC50, 8.57 ± 0.422 μM). A 3D QSAR model [comparative molecular field analysis, q2 = 0.502, optimum number of components (ONC) = 3, R2 = 0.983, F = 404.378, standard error of estimation (SEE) = 0.070, and two descriptors; comparative similarity index analysis, q2 = 0.580, ONC = 10, R2 = 0.999, F = 1617.594, SEE = 0.022, and four descriptors] indicates that the DPP-IV inhibition of flavonoid was facilitated by crucial structural factors. Position 3 of ring C favored bulky, hydrogen bond acceptors and hydrophilic and electron-donating substituents. The presence of minor and electron-withdrawing groups at position 4' of ring B and positions 5 and 7 of ring A could improve DPP-IV inhibition. Moreover, the three flavonoids mentioned above could effectively suppress DPP-IV activity and expression in Caco-2 cells. This work may supply new insights into dietary flavonoids as DPP-IV inhibitors for controlling blood glucose.
Collapse
Affiliation(s)
- Fengyi Gao
- School of Biology and Food, Shangqiu Normal University, Shangqiu, Henan Province 476000, People's Republic of China
| | - Yishan Fu
- Faculty of Agriculture and Food, Yunnan Institute of Food Safety, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China
| | - Junjie Yi
- Faculty of Agriculture and Food, Yunnan Institute of Food Safety, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China
| | - Anning Gao
- School of Biology and Food, Shangqiu Normal University, Shangqiu, Henan Province 476000, People's Republic of China
| | - Yijia Jia
- Faculty of Agriculture and Food, Yunnan Institute of Food Safety, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Shengbao Cai
- Faculty of Agriculture and Food, Yunnan Institute of Food Safety, Kunming University of Science and Technology, Kunming, Yunnan Province 650500, People's Republic of China
| |
Collapse
|
5
|
Hong C, Liang J, Xia J, Zhu Y, Guo Y, Wang A, Lu C, Ren H, Chen C, Li S, Wang D, Zhan H, Wang J. One Stone Four Birds: A Novel Liposomal Delivery System Multi-functionalized with Ginsenoside Rh2 for Tumor Targeting Therapy. NANO-MICRO LETTERS 2020; 12:129. [PMID: 34138128 PMCID: PMC7770862 DOI: 10.1007/s40820-020-00472-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/19/2020] [Indexed: 05/08/2023]
Abstract
Liposomes hold great potential in anti-cancer drug delivery and the targeting treatment of tumors. However, the clinical therapeutic efficacy of liposomes is still limited by the complexity of tumor microenvironment (TME) and the insufficient accumulation in tumor sites. Meanwhile, the application of cholesterol and polyethylene glycol (PEG), which are usually used to prolong the blood circulation and stabilize the structure of liposomes respectively, has been questioned due to various disadvantages. Herein, we developed a ginsenoside Rh2-based multifunctional liposome system (Rh2-lipo) to effectively address these challenges once for all. Different with the conventional 'wooden' liposomes, Rh2-lipo is a much more brilliant carrier with multiple functions. In Rh2-lipo, both cholesterol and PEG were substituted by Rh2, which works as membrane stabilizer, long-circulating stealther, active targeting ligand, and chemotherapy adjuvant at the same time. Firstly, Rh2 could keep the stability of liposomes and avoid the shortcomings caused by cholesterol. Secondly, Rh2-lipo showed a specifically prolonged circulation behavior in the blood. Thirdly, the accumulation of the liposomes in the tumor was significantly enhanced by the interaction of glucose transporter of tumor cells with Rh2. Fourth, Rh2-lipo could remodel the structure and reverse the immunosuppressive environment in TME. When tested in a 4T1 breast carcinoma xenograft model, the paclitaxel-loaded Rh2-lipo realized high efficient tumor growth suppression. Therefore, Rh2-lipo not only innovatively challenges the position of cholesterol as a liposome component, but also provides another innovative potential system with multiple functions for anti-cancer drug delivery.
Collapse
Affiliation(s)
- Chao Hong
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Jianming Liang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Ying Zhu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Yizhen Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Anni Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Chunyi Lu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Hongwei Ren
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Chen Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Shiyi Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
| | - Dan Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China
- Shanghai Ginposome Pharmatech Co., Ltd, Shanghai, 201600, People's Republic of China
| | - Huaxing Zhan
- Shanghai Ginposome Pharmatech Co., Ltd, Shanghai, 201600, People's Republic of China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, People's Republic of China.
- Institute of Integrated Chinese and Western Medicine, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
6
|
Sharma S, Tripathi P, Sharma J, Dixit A. Flavonoids modulate tight junction barrier functions in hyperglycemic human intestinal Caco-2 cells. Nutrition 2020; 78:110792. [PMID: 32473529 DOI: 10.1016/j.nut.2020.110792] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/01/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Diabetes mellitus is a chronic disease requiring lifelong medical attention. With hundreds of millions suffering worldwide and a rapidly rising incidence, diabetes mellitus poses a great burden on health care systems. Recent studies investigating the underlying mechanisms involved in disease development in diabetes point to the role of the dysregulation of the intestinal barrier. Hyperglycemia-mediated tight junction deformity is known to contribute to leaky gut in various metabolic disorders. The present study aimed to investigate the role of oxidative stress on intestinal epithelial tight junction (TJ) barrier functions in hyperglycemia. Because many flavonoids are known to influence the cellular redox state, exploring these flavonoids may help to understand the role of TJ barrier in hyperglycemia-mediated oxidative stress, which in turn might unfold the association of oxidative stress and dysfunction of barrier-forming TJs. METHODS Caco-2 cells were stimulated with high glucose (HG), with or without flavonoids (quercetin, morin, naringenin), for 24 h. We determined cellular viability, levels of reactive oxygen species, and mitochondrial membrane potential in flavonoids treated HG-Caco-2 cells. The levels of the proinflammatory cytokines, glucose uptake, and expression of glucose transporters were determined on flavonoids treatment. We investigated the effect of flavonoids on TJs functions by measuring transepithelial electrical resistance (a TJ integrity marker), membrane permeability using tracer compounds, and the expressions levels of TJs related molecules on hyperglycemic Caco-2 cell monolayers. RESULTS We found that high glucose treatment resulted in reduced cell viability, increased reactive oxygen species production, measurable mitochondrial dysfunction, and decreased transepithelial electrical resistance, with increased membrane permeability. Treatment with the test flavonoids produced increased cell viability and reduced glucose uptake of HG-Caco-2 cells. A concomitant decrease in reactive oxygen species production, proinflammatory cytokines, and Glut-associated genes and proteins were identified with flavonoid treatment. Flavonoids prevented derangement of TJs protein interaction and stabilized membrane permeability. CONCLUSIONS These findings indicate that flavonoids confer protection against hyperglycemia-mediated oxidative stress and enhance intestinal barrier functions by modulating underlying intracellular molecular mechanisms.
Collapse
Affiliation(s)
- Sapna Sharma
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Prabhanshu Tripathi
- Translational Health Science, and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Jeetesh Sharma
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Aparna Dixit
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
7
|
Ginsenoside Rg1 and the control of inflammation implications for the therapy of type 2 diabetes: A review of scientific findings and call for further research. Pharmacol Res 2020; 152:104630. [DOI: 10.1016/j.phrs.2020.104630] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/30/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
|
8
|
Fujiwara M, Kuwahara D, Hayashi M, Zendo T, Sato M, Nakayama J, Sonomoto K. Lowering effect of viable Pediococcus pentosaceus QU 19 on the rise in postprandial glucose. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2020; 39:57-64. [PMID: 32328401 PMCID: PMC7162691 DOI: 10.12938/bmfh.19-041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/31/2019] [Indexed: 01/31/2023]
Abstract
In the present study, we investigated the glucose-decreasing action of lactic acid bacteria (LAB). The finding of this study could be helpful for people in controlling their blood sugar
levels. The LAB candidate was isolated from a Japanese fermented food and identified as Pediococcus pentosaceus by an analysis of its genome sequence. Postprandial blood
glucose elevation was investigated using oral starch tolerance tests in mice. Normal mice were fed starch and lyophilized cells of P. pentosaceus QU 19 at the same time.
Even without pre-administration of P. pentosaceus QU 19, elevation of the blood glucose level was significantly suppressed by the intake of P. pentosaceus
QU 19 at the same time as oral administration of starch. According to the results for its survival in simulated digestive juice and the reduction of blood glucose level in mice, P.
pentosaceus QU 19 has potential hypoglycemic activity. In vitro measurements revealed that the glucose-decreasing action of P. pentosaceus QU 19
is probably caused by the glucose assimilation of the strain, not the inhibition of carbohydrate-splitting enzymes which has been reported for other LABs previously. These findings indicate
that specific strains of LAB, especially P. pentosaceus QU 19, and foods fermented by LAB may be beneficial for people who must manage glucose ingestion.
Collapse
Affiliation(s)
- Miki Fujiwara
- Biotechnology R&D Group, HPM Research & Development Department, High Performance Materials Company, JXTG Nippon Oil & Energy Corporation, 8 Chidoricho, Naka-ku, Yokohama 231-0815, Japan.,Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Daichi Kuwahara
- Biotechnology R&D Group, HPM Research & Development Department, High Performance Materials Company, JXTG Nippon Oil & Energy Corporation, 8 Chidoricho, Naka-ku, Yokohama 231-0815, Japan
| | - Masahiro Hayashi
- Biotechnology R&D Group, HPM Research & Development Department, High Performance Materials Company, JXTG Nippon Oil & Energy Corporation, 8 Chidoricho, Naka-ku, Yokohama 231-0815, Japan
| | - Takeshi Zendo
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masao Sato
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Jiro Nakayama
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kenji Sonomoto
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
9
|
Chaunchaiyakul R, Leelayuwat N, Wu JF, Huang CY, Kuo CH. Contrasting actions of ginsenosides Rb1 and Rg1 on glucose tolerance in rats. CHINESE J PHYSIOL 2019; 62:267-272. [PMID: 31793463 DOI: 10.4103/cjp.cjp_61_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ginsenoside profile of Panax ginseng is changing with season and cultivated soil. Yet, dose-response relationship of main ginsenosides on metabolic measures has not been documented in vivo. Here, we examined glucose and insulin responses after an oral glucose challenge (0.5 g/kg body weight) at various doses (0.01, 0.1, 1, and 10 mg/kg of body weight) under acute and chronic Rb1 and Rg1 supplemented conditions. The results show that Rb1 (0.01 and 0.1 mg/kg body weight) increased, whereas Rg1 (0.01 mg/kg body weight) decreased postprandial glucose levels compared with the Vehicle group (P < 0.05). This contrasting effect reduced as dose increased. Both Rb1 and Rg1 decreased the mitochondrial enzyme citrate synthase activity (P < 0.05) together with decreases in glycogen content in red gastrocnemius muscle and body temperature at low doses (P < 0.05), compared with the Vehicle group. These differences also diminished as dosage increases. For reliable ginseng research, dose standardization on Rg1 and Rb1 is essential based on their opposing action and peculiar dose-response relationship. Both major ginsenosides may influence dynamics of mitochondria turnover and alter muscle metabolism.
Collapse
Affiliation(s)
- Rungchai Chaunchaiyakul
- Faculty of Sports Science and Technology, Mahidol University, Nakhonpathom, Thailand; Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Naruemon Leelayuwat
- Exercise and Sport Sciences Development and Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jin-Fu Wu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education & Sports Science, South China Normal University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| |
Collapse
|
10
|
Toxicological Evaluation of a Mixture of Astragalus membranaceus and Panax notoginseng Root Extracts (InnoSlim®). J Toxicol 2019; 2019:5723851. [PMID: 31354815 PMCID: PMC6633876 DOI: 10.1155/2019/5723851] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/16/2022] Open
Abstract
Astragalus spp. and Panax spp. have a long history of traditional human use. A blend, InnoSlim®, of highly purified and fractionated root extracts from Astragalus membranaceus and Panax notoginseng has now been developed for human consumption; however, the unique constituent content of this blend has not been specifically evaluated with respect to safety. Therefore, the toxicological potential of the blend was formally investigated in a series of studies—genetic toxicity was evaluated in a bacterial reverse mutation test followed by an in vivo mammalian micronucleus test, and general toxicity was evaluated in a 28-day repeated-dose oral toxicity study in rats. No evidence of mutagenicity was observed in the bacterial tester strains used, and no evidence of in vivo chromosomal damage resulting in increased frequency of micronucleated cells was observed in male Crl:NMRI BR mice. No mortality or toxic effects were observed, and no target organs were identified, in male and female Han:WIST rats exposed to 0, 400, 800, or 1200 mg/kg bw/day of the blend by gavage for 28 consecutive days. The highest dose—1200 mg/kg bw/day—was determined to be the NOAEL. Based on these results, extrapolation towards a safe human consumption level can be explored.
Collapse
|
11
|
Intestinal Saturated Long-Chain Fatty Acid, Glucose and Fructose Transporters and Their Inhibition by Natural Plant Extracts in Caco-2 Cells. Molecules 2018; 23:molecules23102544. [PMID: 30301205 PMCID: PMC6222386 DOI: 10.3390/molecules23102544] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/29/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023] Open
Abstract
The intestinal absorption of fatty acids, glucose and fructose is part of the basic requirements for the provision of energy in the body. High access of saturated long-chain fatty acids (LCFA), glucose and fructose can facilitate the development of metabolic diseases, particularly the metabolic syndrome and type-2 diabetes mellitus (T2DM). Research has been done to find substances which decelerate or inhibit intestinal resorption of these specific food components. Promising targets are the inhibition of intestinal long-chain fatty acid (FATP2, FATP4), glucose (SGLT1, GLUT2) and fructose (GLUT2, GLUT5) transporters by plant extracts and by pure substances. The largest part of active components in plant extracts belongs to the group of polyphenols. This review summarizes the knowledge about binding sites of named transporters and lists the plant extracts which were tested in Caco-2 cells regarding uptake inhibition.
Collapse
|
12
|
Hong C, Yang P, Li S, Guo Y, Wang D, Wang J. In Vitro/In Vivo Metabolism of Ginsenoside Rg5 in Rat Using Ultra-Performance Liquid Chromatography/Quadrupole-Time-of-Flight Mass Spectrometry. Molecules 2018; 23:E2113. [PMID: 30135411 PMCID: PMC6225384 DOI: 10.3390/molecules23092113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/02/2018] [Accepted: 08/05/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Ginsenoside Rg5 has been proved to have a wide range of pharmacological activities. However, the in vitro and in vivo metabolism pathways of ginsenosides are still unclear, which impedes the understanding of their in vivo fate. In this paper, the possible metabolic process of Rg5 was studied and the metabolites are identified. Methods: Samples from rat liver microsomes (RLMs) in vitro and from rat urine, plasma and feces in vivo were collected for analysis after oral administration of Rg5. A rapid analysis technique using ultra-performance liquid chromatography (UPLC)/quadrupole-time-of-flight mass spectrometry (QTOF-MS) was applied for detecting metabolites of Rg5 both in vitro and in vivo. Results: A feasible metabolic pathway was proposed and described for ginsenoside Rg5. A total of 17 metabolic products were detected in biological samples, including the RLMs (four), rat urine (two), feces (13) and plasma (four). Fifteen of them have never been reported before. Oxidation, deglycosylation, deoxidation, glucuronidation, demethylation and dehydration were found to be the major metabolic reactions of Rg5. Conclusions: The present study utilized a reliable and quick analytical tool to explore the metabolism of Rg5 in rats and provided significant insights into the understanding of the metabolic pathways of Rg5 in vitro and in vivo. The results could be used to not only evaluate the efficacy and safety of Rg5, but also identify potential active drug candidates from the metabolites.
Collapse
Affiliation(s)
- Chao Hong
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| | - Ping Yang
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Shuping Li
- The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China.
| | - Yizhen Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| | - Dan Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
- Institute of Materia Medica, Academy of Integrated Chinese and Western Medicine, Fudan University, Shanghai 200040, China.
| |
Collapse
|
13
|
Müller U, Stübl F, Schwarzinger B, Sandner G, Iken M, Himmelsbach M, Schwarzinger C, Ollinger N, Stadlbauer V, Höglinger O, Kühne T, Lanzerstorfer P, Weghuber J. In Vitro and In Vivo Inhibition of Intestinal Glucose Transport by Guava (Psidium Guajava) Extracts. Mol Nutr Food Res 2018; 62:e1701012. [PMID: 29688623 PMCID: PMC6001447 DOI: 10.1002/mnfr.201701012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/15/2018] [Indexed: 12/21/2022]
Abstract
SCOPE Known pharmacological activities of guava (Psidium guajava) include modulation of blood glucose levels. However, mechanistic details remain unclear in many cases. METHODS AND RESULTS This study investigated the effects of different guava leaf and fruit extracts on intestinal glucose transport in vitro and on postprandial glucose levels in vivo. Substantial dose- and time-dependent glucose transport inhibition (up to 80%) was observed for both guava fruit and leaf extracts, at conceivable physiological concentrations in Caco-2 cells. Using sodium-containing (both glucose transporters, sodium-dependent glucose transporter 1 [SGLT1] and glucose transporter 2 [GLUT2], are active) and sodium-free (only GLUT2 is active) conditions, we show that inhibition of GLUT2 was greater than that of SGLT1. Inhibitory properties of guava extracts also remained stable after digestive juice treatment, indicating a good chemical stability of the active substances. Furthermore, we could unequivocally show that guava extracts significantly reduced blood glucose levels (≈fourfold reduction) in a time-dependent manner in vivo (C57BL/6N mice). Extracts were characterized with respect to their main putative bioactive compounds (polyphenols) using HPLC and LC-MS. CONCLUSION The data demonstrated that guava leaf and fruit extracts can potentially contribute to the regulation of blood glucose levels.
Collapse
Affiliation(s)
- Ulrike Müller
- University of Applied Sciences Upper Austria4600WelsAustria
| | - Flora Stübl
- University of Applied Sciences Upper Austria4600WelsAustria
| | - Bettina Schwarzinger
- University of Applied Sciences Upper Austria4600WelsAustria
- Austrian Competence Center for Feed and Food QualitySafety and Innovation4600WelsAustria
| | - Georg Sandner
- University of Applied Sciences Upper Austria4600WelsAustria
| | | | - Markus Himmelsbach
- Johannes Kepler UniversityInstitute for Analytical Chemistry4040LinzAustria
| | - Clemens Schwarzinger
- Johannes Kepler UniversityInstitute for Chemical Technology of Organic Materials4040LinzAustria
| | - Nicole Ollinger
- University of Applied Sciences Upper Austria4600WelsAustria
- Austrian Competence Center for Feed and Food QualitySafety and Innovation4600WelsAustria
| | - Verena Stadlbauer
- University of Applied Sciences Upper Austria4600WelsAustria
- Austrian Competence Center for Feed and Food QualitySafety and Innovation4600WelsAustria
| | | | | | | | - Julian Weghuber
- University of Applied Sciences Upper Austria4600WelsAustria
- Austrian Competence Center for Feed and Food QualitySafety and Innovation4600WelsAustria
| |
Collapse
|
14
|
Oat globulin peptides regulate antidiabetic drug targets and glucose transporters in Caco-2 cells. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.061] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
15
|
Abstract
Although ginseng has been shown to have an antiobesity effect, antiobesity-related mechanisms are complex and have not been completely elucidated. In the present study, we evaluated ginseng’s effects on food intake, the digestion, and absorption systems, as well as liver, adipose tissue, and skeletal muscle in order to identify the mechanisms involved. A review of previous in vitro and in vivo studies revealed that ginseng and ginsenosides can increase energy expenditure by stimulating the adenosine monophosphate-activated kinase pathway and can reduce energy intake. Moreover, in high fat diet-induced obese and diabetic individuals, ginseng has shown a two-way adjustment effect on adipogenesis. Nevertheless, most of the previous studies into antiobesity effects of ginseng have been animal based, and there is a paucity of evidence supporting the suggestion that ginseng can exert an antiobesity effect in humans.
Collapse
|
16
|
Gao S, Kushida H, Makino T. Ginsenosides, ingredients of the root of Panax ginseng, are not substrates but inhibitors of sodium-glucose transporter 1. J Nat Med 2016; 71:131-138. [DOI: 10.1007/s11418-016-1042-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/31/2016] [Indexed: 02/07/2023]
|
17
|
Wang CW, Su SC, Huang SF, Huang YC, Chan FN, Kuo YH, Hung MW, Lin HC, Chang WL, Chang TC. An Essential Role of cAMP Response Element Binding Protein in Ginsenoside Rg1-Mediated Inhibition of Na+/Glucose Cotransporter 1 Gene Expression. Mol Pharmacol 2015; 88:1072-83. [PMID: 26429938 DOI: 10.1124/mol.114.097352] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 09/23/2015] [Indexed: 02/14/2025] Open
Abstract
The Na(+)/glucose cotransporter 1 (SGLT1) is responsible for glucose uptake in intestinal epithelial cells. It has been shown that the intestinal SGLT1 level is significantly increased in diabetic individuals and positively correlated with the pathogenesis of diabetes. The development of targeted therapeutics that can reduce the intestinal SGLT1 expression level is, therefore, important. In this study, we showed that ginsenoside Rg1 effectively decreased intestinal glucose uptake through inhibition of SGLT1 gene expression in vivo and in vitro. Transient transfection analysis of the SGLT1 promoter revealed an essential cAMP response element (CRE) that confers the Rg1-mediated inhibition of SGLT1 gene expression. Chromatin immunoprecipitation assay and targeted CRE-binding protein (CREB) silencing demonstrated that Rg1 reduced the promoter binding of CREB and CREB binding protein associated with an inactivated chromatin status. In addition, further studies showed that the epidermal growth factor receptor (EGFR) signaling pathway also plays an essential role in the inhibitory effect of Rg1; taken together, our study demonstrates the involvement of the EGFR-CREB signaling pathway in the Rg1-mediated downregulation of SGLT1 expression, which offers a potential strategy in the development of antihyperglycemic and antidiabetic treatments.
Collapse
Affiliation(s)
- Chun-Wen Wang
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Shih-Chieh Su
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Shu-Fen Huang
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Yu-Chuan Huang
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Fang-Na Chan
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Yu-Han Kuo
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Mei-Whey Hung
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Hang-Chin Lin
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.)
| | - Wen-Liang Chang
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.).
| | - Tsu-Chung Chang
- Graduate Institute of Life Sciences (C.-W.W., T.-C.C.), Department of Biochemistry (S.-C.S., S.-F.H., F.-N.C., Y.-H.K., T.-C.C.), Institute of Preventive Medicine (Y.-C.H.), and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, Republic of China (H.-C.L., W.-L.C.); Department of Research and Education, Veteran General Hospital, Taipei, Taiwan, Republic of China (M.-W.H.); Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, Republic of China (T.-C.C.); and Department of Biotechnology, Asia University, Taichung, Taiwan, Republic of China (T.-C.C.).
| |
Collapse
|
18
|
Chang WH, Tsai YL, Huang CY, Hsieh CC, Chaunchaiyakul R, Fang Y, Lee SD, Kuo CH. Null effect of ginsenoside Rb1 on improving glycemic status in men during a resistance training recovery. J Int Soc Sports Nutr 2015; 12:34. [PMID: 26300710 PMCID: PMC4545376 DOI: 10.1186/s12970-015-0095-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 08/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ginsenoside Rb1, a principle active ingredients of Panax ginseng, has been shown to lower blood glucose in animals and increase insulin secretion in cultured insulinoma cells. The aim of this study was to determine the effects of daily ginsenoside Rb1 supplementation on circulating glucose and insulin levels in men during a 5-day recovery period after an acute bout of resistance exercise. METHODS Twelve gymnasts (20.5 ± 0.3 years of age) participated in this double blind placebo-controlled crossover trial. They were challenged by a lower-limb resistance exercise at a weight load of 85 % one-repetition maximal (1-RM) for 10 repetitions, six sets of the movement. Rb1 (1 ng/kg) or Placebo was orally delivered to participants daily during a 5-day recovery period after challenge. Circulating insulin, glucose and heart rate variability (HRV) were measured under fasted condition in the morning at Days 1, Day 3, and Day 5 during recovery. RESULTS No significant effect of Rb1 on circulating glucose and insulin levels were found among participants during the 5-day recovery period. A persistent elevation in sympathetic nervous activity, indicated by increased HRV-low frequency/high frequency (HRV-LF/HF) power, during the Rb1 trial was observed. CONCLUSIONS The result of the study suggests that the null effect of Rb1 supplementation on lowering glucose and insulin levels of participants may be associated with chronic sympathetic activation.
Collapse
Affiliation(s)
- Wei-Hsiang Chang
- Department of Sports Sciences, Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan.,Department of Physical Education, National Hsinchu University of Education, Hsinchu, Taiwan.,Department of Athletic Training and Health, National Taiwan Sport University, Taoyuan, Taiwan
| | - Ying-Lan Tsai
- Department of Physical Education, National Hsinchu University of Education, Hsinchu, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - City C Hsieh
- Department of Physical Education, National Hsinchu University of Education, Hsinchu, Taiwan
| | | | - Yu Fang
- Department of Sports Sciences, Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Shin-Da Lee
- Department of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan.,Department of Rehabilitation Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
19
|
Wang CW, Chang WL, Huang YC, Chou FC, Chan FN, Su SC, Huang SF, Ko HH, Ko YL, Lin HC, Chang TC. An essential role of cAMP response element-binding protein in epidermal growth factor-mediated induction of sodium/glucose cotransporter 1 gene expression and intestinal glucose uptake. Int J Biochem Cell Biol 2015; 64:239-51. [PMID: 25936754 DOI: 10.1016/j.biocel.2015.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/26/2015] [Accepted: 04/13/2015] [Indexed: 02/06/2023]
Abstract
The sodium/glucose cotransporter 1 (SGLT1) is responsible for glucose uptake in intestinal epithelial cells. Its expression is decreased in individuals with intestinal inflammatory disorders and is correlated with the pathogenesis of disease. The aim of this study was to understand the regulatory mechanism of the SGLT1 gene. Using the trinitrobenzene sulfonic acid-induced mouse models of intestinal inflammation, we observed decreased SGLT1 expression in the inflamed intestine was positively correlated with the mucosal level of epidermal growth factor (EGF) and activated CREB. Overexpression of EGF demonstrated that the effect of EGF on intestinal glucose uptake was primarily due to the increased level of SGLT1. We identified an essential cAMP binding element (CRE) confers EGF inducibility in the human SGLT1 gene promoter. ChIP assay further demonstrated the increased binding of CREB and CBP to the SGLT1 gene promoter in EGF-treated cells. In addition, the EGFR- and PI3K-dependent CREB phosphorylations are involved in the EGF-mediated SGLT1 expression. This is the first report to demonstrate that CREB is involved in EGF-mediated transcription regulation of SGLT1 gene in the normal and inflamed intestine, which can provide potential therapeutic applications for intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Chun-Wen Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Wen-Liang Chang
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yu-Chuan Huang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Fang-Chi Chou
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Fang-Na Chan
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shih-Chieh Su
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shu-Fen Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hui-Hsuan Ko
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-Ling Ko
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hang-Chin Lin
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Tsu-Chung Chang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
20
|
Wang CW, Huang YC, Chan FN, Su SC, Kuo YH, Huang SF, Hung MW, Lin HC, Chang WL, Chang TC. A gut microbial metabolite of ginsenosides, compound K, induces intestinal glucose absorption and Na(+) /glucose cotransporter 1 gene expression through activation of cAMP response element binding protein. Mol Nutr Food Res 2015; 59:670-84. [PMID: 25600494 DOI: 10.1002/mnfr.201400688] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/13/2014] [Accepted: 12/30/2014] [Indexed: 01/21/2023]
Abstract
SCOPE The Na(+) /glucose cotransporter 1 (SGLT1) plays a crucial role in glucose uptake in intestinal epithelial cells (IECs), which has been shown essential in ameliorating intestinal inflammation. Ginseng has historically been used to treat inflammatory disorders. Understanding the regulatory mechanism of ginseng-mediated induction of SGLT1 gene expression in human intestinal cells is therefore important. METHODS AND RESULTS We demonstrate that ginsenoside compound K (CK) enhances SGLT1-mediated glucose uptake in mice and human intestinal Caco-2 cells. Transient transfection analysis using SGLT1 promoter-luciferase reporters demonstrated that the presence of an essential cAMP response element (CRE) is required for CK-mediated induction of SGLT1 gene expression. The ChIP assays indicated that increased CRE-binding protein (CREB) and CREB-binding protein (CBP) binding to the SGLT1 promoter in CK-treated cells is associated with an activated chromatin state. Our result showed that the increased CREB phosphorylation is directly correlated with SGLT1 expression in IECs. Further studies indicated that the epidermal growth factor receptor (EGFR) signaling pathway is involved in the CK-mediated effect. CONCLUSION These findings provide a novel mechanism for the CK-mediated upregulation of SGLT1 expression through EGFR-CREB signaling activation, which could contribute to reducing gut inflammation.
Collapse
Affiliation(s)
- Chun-Wen Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Li KK, Gong XJ. A review on the medicinal potential of Panax ginseng saponins in diabetes mellitus. RSC Adv 2015. [DOI: 10.1039/c5ra05864c] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This review article summarizes the anti-diabetic effects and mechanisms ofPanax ginsengsaponins and its active specific ginsenosides.
Collapse
Affiliation(s)
- Ke-Ke Li
- School of Medical
- Dalian University
- Dalian 116622
- P. R. China
| | - Xiao-Jie Gong
- School of Medical
- Dalian University
- Dalian 116622
- P. R. China
| |
Collapse
|
22
|
Effects of Food Lectins on the Transport System of Human Intestinal Caco-2 Cell Monolayers. Biosci Biotechnol Biochem 2014; 77:1917-24. [DOI: 10.1271/bbb.130367] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Talukder JR, Griffin A, Jaima A, Boyd B, Wright J. Lactoferrin ameliorates prostaglandin E2-mediated inhibition of Na+-glucose cotransport in enterocytes. Can J Physiol Pharmacol 2014; 92:9-20. [DOI: 10.1139/cjpp-2013-0211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Various immunoinflammatory cytokines are produced during chronic intestinal inflammation, which inhibits Na+-glucose cotransport (SGLT1) in villus cells. Lactoferrin (Lf), abundantly present in colostrum, is a multifunctional glycoprotein that is absorbed by receptor-mediated transcytosis in humans and animals and has been shown to exert anti-inflammatory effects. Therefore, this study aimed to examine whether Lf would prevent PGE2 effect on SGLT1 for glucose absorption in enterocytes. Intestinal epithelial cells (IEC-6) were grown on transwell plates, treated with phlorizin, PGE2, AH6809, and Lf, and 3-O-methyl d-glucopyranose (OMG) uptake was measured in 10 days postconfluent. Na+-dependent OMG uptake, phlorizin, and immunoblotting studies established the activity and apical membrane localization of SGLT1 in IEC-6 cells. PGE2 inhibited SGLT1 in a concentration- and time-dependent manner with an inhibitory constant (Ki) of 50.0 nmol/L and that was antagonized by prostanoid receptor inhibitor, AH6809. PGE2 did not alter Na+/K+-ATPase activity. In contrast, quantitative real-time polymerase chain reaction and Western blot analyses revealed that SGLT1-specific transcripts and protein expression level were decreased 3-fold by PGE2. Furthermore, PGE2 treatment increased intracellular cyclic adenosine monophosphate (cAMP) and Ca2+ concentrations and decreased SGLT1 expression on the apical membrane, and these effects were ameliorated by Lf. Therefore, we conclude that Lf ameliorates the PGE2 inhibition of SGLT1 most likely via the Ca2+- and cAMP-signaling pathways.
Collapse
Affiliation(s)
- Jamilur R. Talukder
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Ashley Griffin
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Antara Jaima
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Brittney Boyd
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| | - Jaleesa Wright
- Department of Biology, LeMoyne-Owen College, 807 Walker Avenue, Memphis, TN 38126, USA
| |
Collapse
|
24
|
Yuan HD, Kim JT, Kim SH, Chung SH. Ginseng and diabetes: the evidences from in vitro, animal and human studies. J Ginseng Res 2013; 36:27-39. [PMID: 23717101 PMCID: PMC3659569 DOI: 10.5142/jgr.2012.36.1.27] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 12/02/2011] [Accepted: 12/02/2011] [Indexed: 01/09/2023] Open
Abstract
Panax ginseng exhibits pleiotropic beneficial effects on cardiovascular system, central nervous system, and immune system. In the last decade, numerous preclinical findings suggest ginseng as a promising therapeutic agent for diabetes prevention and treatment. The mechanism of ginseng and its active components is complex and is demonstrated to either modulate insulin production/secretion, glucose metabolism and uptake, or inflammatory pathway in both insulin-dependent and insulin-independent manners. However, human studies are remained obscure because of contradictory results. While more studies are warranted to further understand these contradictions, ginseng holds promise as a therapeutic agent for diabetes prevention and treatment. This review summarizes the evidences for the therapeutic potential of ginseng and ginsenosides from in vitro studies, animal studies and human clinical trials with a focus on diverse molecular targets including an AMP-activated protein kinase signaling pathway.
Collapse
Affiliation(s)
- Hai-Dan Yuan
- Department of Pharmacology and Clinical Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | |
Collapse
|
25
|
Liu ZQ. Chemical Insights into Ginseng as a Resource for Natural Antioxidants. Chem Rev 2012; 112:3329-55. [DOI: 10.1021/cr100174k] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Zai-Qun Liu
- Department of Organic Chemistry, College
of Chemistry, Jilin University, Changchun
130021, China
| |
Collapse
|
26
|
Faria A, Monteiro R, Pestana D, Freitas VD, Mateus N, Azevedo I, Calhau C. Intestinal oxidative state can alter nutrient and drug bioavailability. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2011; 2:322-7. [PMID: 20716920 PMCID: PMC2835921 DOI: 10.4161/oxim.2.5.9769] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Organic cations (OCs) are substances of endogenous (e.g. dopamine, choline) or exogenous (e.g. drugs like cimetidine) origin that are positively charged at physiological pH. Since many of these compounds can not pass the cell membrane freely, their transport in our out of cells must be mediated by specific transport systems. Transport by organic cation transporters (OCTs) can be regulated rapidly by altering their trafficking and/or affinities in response to a stimuli. However, for example, a specific disease could lead to modifications in the expression of OCTs. Chronic exposure to oxidative stress has been suggested to alter regulation and functional activity of proteins through several pathways. According to results from a previous work, oxidation-reduction pathways were thought to be involved in intestinal organic cation uptake modulation. The present work was performed in order to evaluate the influence of oxidative stressors, especially glutathione, on the intestinal organic cation absorption. For this purpose, the effect of compounds with different redox potential (glutathione, an endogenous antioxidant, and procyanidins, diet antioxidants) was assessed on MPP+ (1-methyl-4-phenylpyridinium iodide) uptake in an enterocyte cell line (Caco-2). Caco-2 cells were subcultured with two different media conditions (physiological: 5 mM glucose, referred as control cells; and high-glucose: 25 mM glucose, referred as HG cells). In HG cells, the uptake was significantly lower than in control cells. Redox changing interventions affected MPP+ uptake, both in control and in high-glucose Caco-2 cells. Cellular glutathione levels could have an important impact on membrane transporters activity. The results indicate that modifications in the cellular oxidative state modulate MPP+ uptake by Caco-2 cells. Such modifications may reflect in changes of nutrient and drug bioavailability.
Collapse
Affiliation(s)
- Ana Faria
- University of Porto, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
27
|
Hao H, Lai L, Zheng C, Wang Q, Yu G, Zhou X, Wu L, Gong P, Wang G. Microsomal cytochrome p450-mediated metabolism of protopanaxatriol ginsenosides: metabolite profile, reaction phenotyping, and structure-metabolism relationship. Drug Metab Dispos 2010; 38:1731-9. [PMID: 20639434 DOI: 10.1124/dmd.110.033845] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Although the biotransformation of ginsenosides in the gastrointestinal tract has been extensively studied, much less is known about hepatic cytochrome P450 (P450)-catalyzed metabolism. The major aims of this study were to clarify the metabolic pathway and P450 isoforms involved and to explore the structure-metabolism relationship of protopanaxatriol (PPT)-type ginsenosides in hepatic microsomes. Efficient depletion of ginsenoside Rh1, Rg2, Rf, and PPT was found, whereas the elimination of Re and Rg1, characterized by a glucose substitution at the C20 hydroxy group, was negligible in microsomal incubation systems. Based on high-performance liquid chromatography hybrid ion trap and time-of-flight mass spectrometry analysis, the oxygenation metabolism on the C20 aliphatic branch chain was identified as the predominant metabolic pathway of PPT ginsenosides in both human and rat hepatic microsomes. By a comparison with authentic standards, the C24-25 double bond was identified as one of the oxygenation sites to produce the metabolites of C20-24 epoxide (ocotillol-type ginsenosides). Both chemical inhibition and human recombinant P450 isoform assays indicated that CYP3A4 was the predominant isozyme responsible for the oxygenation metabolism of PPT ginsenosides. Enzyme kinetic evaluations in rat and human hepatic microsomes and human recombinant CYP3A4 isozyme incubation systems showed generally consistent results in that the intrinsic clearance ranked as Rf ≤ Rg2 < Rh1 < PPT, closely correlating with logP values and the number of glycosyl substitutions. Results obtained from this study suggest that CYP3A4-catalyzed oxygenation metabolism plays an important role in the hepatic disposition of ginsenosides and that glycosyl substitution, especially at the C20 hydroxy group, determines their intrinsic clearances by CYP3A4.
Collapse
Affiliation(s)
- Haiping Hao
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kim SJ, Yuan HD, Chung SH. Ginsenoside Rg1 suppresses hepatic glucose production via AMP-activated protein kinase in HepG2 cells. Biol Pharm Bull 2010; 33:325-8. [PMID: 20118562 DOI: 10.1248/bpb.33.325] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Panax ginseng is known to have anti-diabetic activity, but the active ingredients are not yet fully identified. In this study, we found the inhibitory effect of Rg(1) on hepatic glucose production through AMP-activated protein kinase (AMPK) activation in HepG2 cells. Rg(1) significantly inhibited hepatic glucose production in a concentration-dependent manner, and this effect was reversed in the presence of compound C, a selective AMPK inhibitor. In addition, Rg(1) markedly induced the phosphorylations of liver kinase B1 (LKB1), AMPK and forkhead box class O1 (FoxO1), a key transcription factor for gluconeogenic enzymes, in time- and concentration-dependent manners. Glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) activities were inhibited by 24% and 21%, respectively, when the cells were exposed to 40 microM of Rg(1), resulting from phosphorylation of FoxO1 and inhibition of gluconeogenic gene expression. Taken together, our results demonstrated the suppressive effect of Rg(1) on hepatic glucose production via LKB1-AMPK-FoxO1 pathway in HepG2 human hepatoma cells.
Collapse
Affiliation(s)
- Sung Jip Kim
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | |
Collapse
|
29
|
Metabolism of Ginsenosides to Bioactive Compounds by Intestinal Microflora and Its Industrial Application. J Ginseng Res 2009. [DOI: 10.5142/jgr.2009.33.3.165] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
30
|
The protective effects of ginsenosides on human erythrocytes against hemin-induced hemolysis. Food Chem Toxicol 2008; 46:886-92. [DOI: 10.1016/j.fct.2007.10.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 09/10/2007] [Accepted: 10/09/2007] [Indexed: 11/24/2022]
|