101
|
Li X, Huang F, Zhu L, Luo T, Zhang Y, Gu H, Guo L, Mao S. Effects of combination therapy with Shenfu Injection in critically ill patients with septic shock receiving mechanical ventilation: A multicentric, real-world study. Front Pharmacol 2022; 13:1041326. [PMID: 36438846 PMCID: PMC9682251 DOI: 10.3389/fphar.2022.1041326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/26/2022] [Indexed: 10/19/2024] Open
Abstract
Background: Septic shock has increasingly become a cause of death threatening human survival. Shenfu Injection (SFI), a patented Chinese medicine, has been widely used in the treatment of patients with sepsis and cardiovascular diseases domestically. We sought to examine whether combination therapy with SFI can improve clinical outcomes in critically ill patients undergoing mechanical ventilation (MV). Methods: This real-world, multicenter retrospective trial enrolled consecutive adult patients with sepsis requiring MV from four medical/surgical intensive care units (ICUs) in China between August 2016 and September 2021. Patients were identified from the medical information department database of each center and assigned to either of two groups (SFI or control) on the basis of the initial treatment received. The primary outcome was 28-day all-cause mortality, and the durations of vasopressor therapy and MV, the ICU length of stay, and costs were assessed as secondary outcomes. Subsequently, we performed a meta-analysis of randomized controlled trials (RCTs) on SFI published before July 2021 to verify our conclusions. Results: 2311 mechanically ventilated patients with septic shock (1128 patients in the SFI group and 1183 in the control group) were analyzed. The survival probability during the first 28 days after admission in the SFI group was greater than that in the control group [p < 0.01 by log-rank test; hazard ratio (HR), 0.56; 95% confidence interval (CI), 0.39-0.72]. Patients in the SFI group also experienced a significantly reduced duration of vasopressor therapy [7.28 (95% CI, 6.14-8.42) vs. 12.06 (95% CI, 10.71-13.41) days, p < 0.001], more ventilator-free days [6.49 (95% CI, 5.42-7.55) vs. 10.84 (95% CI, 9.59-12.09) days, p < 0.001], a shorter ICU length of stay [18.48 (95% CI, 17.59-19.38) vs. 23.77 (95% CI, 22.47-25.07) days, p < 0.001], and more time free from organ failure [14.23 (95% CI, 12.94-15.52) vs. 19.07 (95% CI, 16.09-22.05) days, p < 0.001]. No major adverse effects were reported in either group. Conclusion: Among critically ill patients requiring MV, combination therapy with SFI can improve the survival probability without any obvious adverse reactions.
Collapse
Affiliation(s)
- Xiaoqian Li
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fan Huang
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Lixia Zhu
- Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, China
| | - Tianyi Luo
- Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, China
| | - Yuzhuo Zhang
- Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, China
| | - Huiwen Gu
- Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, China
| | - Liheng Guo
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuai Mao
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
102
|
Shen K, Wu D, Sun B, Zhu Y, Wang H, Zou W, Ma Y, Lu Z. Ginsenoside Rg1 promotes astrocyte-to-neuron transdifferentiation in rat and its possible mechanism. CNS Neurosci Ther 2022; 29:256-269. [PMID: 36352836 PMCID: PMC9804042 DOI: 10.1111/cns.14000] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Neuronal loss caused by spinal cord injury (SCI) usually contributes to irreversible motor dysfunction. Promoting neuronal regeneration and functional recovery is vital to the repair of SCI. AIMS Astrocytes, activated by SCI with high proliferative capacity and proximity to neuronal lineage, are considered ideal cells for neuronal regeneration. As previous studies identified several small molecules for the induction of astrocyte-to-neuron, we confirmed that ginsenoside Rg1, a neuroprotective herb, could promote the direct transdifferentiation of astrocyte-to-neuron in rat. METHODS AND RESULTS Immunofluorescence staining showed that 26.0 ± 1.5% of the induced cells exhibited less astroglial properties and more neuronal markers with typical neuronal morphologies, reflecting 20.6 ± 0.9% of cholinergic neurons and 22.3 ± 1.9% of dopaminergic neurons. Western blot and qRT-PCR revealed that the induced cells had better antiapoptotic ability and Rg1-promoted neuronal transdifferentiation of reactive astrocytes might take effect through suppressing Notch/Stat3 signal pathway. In vivo, a revised SCI model treated by Rg1 was confirmed with faster functional recovery and less injury lesion cavity. CONCLUSION In summary, our study provided a novel strategy of direct transdifferentiation of endogenous rat reactive astrocytes into neurons with Rg1 and promotion of neuronal regeneration after SCI.
Collapse
Affiliation(s)
- Kelv Shen
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Duanrong Wu
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Baihan Sun
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Yin Zhu
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Hao Wang
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Wenjun Zou
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Yuhang Ma
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| | - Zhengfeng Lu
- Department of OrthopedicsThe Second Affliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
103
|
Lee N, Lee JH, Won JE, Lee YJ, Hyun SH, Yi YD, In G, Han HD, Lee Y. KRG and its major ginsenosides do not show distinct steroidogenic activities examined by the OECD test guideline 440 and 456 assays. J Ginseng Res 2022; 47:385-389. [DOI: 10.1016/j.jgr.2022.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 03/12/2023] Open
|
104
|
Baranauskaite J, Ockun MA, Uner B, Gungor B, Duman G, Tas C, Yesilada E. Development and In vitro characterization of pullulan fast dissolving films loaded with Panax ginseng extract, antioxidant properties and cytotoxic efficiency on lung and breast cancer cell lines. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
105
|
Zhang Z, Zhang Z, Yu Z, Chen S, Zhang M, Zhang T, Luo X, Zhao J, Li Z. Simultaneous Improvement of Final Product-Tolerance and Thermostability of GH39 Xylosidase for Prebiotic Production by Directed Evolution. Foods 2022; 11:foods11193039. [PMID: 36230114 PMCID: PMC9563585 DOI: 10.3390/foods11193039] [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: 08/28/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
Xylosidases are widely used for the production of prebiotics and the transformation of natural active substances in the food industry. However, xylosidases with excellent thermostability and product tolerance are required for industrial applications. In this study, the thermostability and final-product tolerance of the previously reported robust xylosidase Xyl21 were further improved via directed evolution. The triple mutant variant Xyl21-A16 (K16R, L94I, and K262N) showed significantly enhanced xylose tolerance, ethanol tolerance, and thermostability with no apparent changes in the specific activity, optimum pH, and temperature compared with the wild type. Single site mutations suggested that variant Xyl21-A16 is the cumulative result of three mutated sites, which indicated that K16 and L94 play important roles in enzyme characteristics. Moreover, a comparison of the predicted protein structures of Xyl21 and its variant indicated that additional molecular interactions formed by K16R and K262N might directly improve the rigidity of the protein structure, therefore contributing to the increased thermostability and product tolerance. The variant Xyl21-A16 developed in this study has great application potential in the production of prebiotics, and also provides a useful reference for the future engineering of other xylosidases.
Collapse
Affiliation(s)
- Zirui Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhengjie Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhao Yu
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shiheng Chen
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Mengwei Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tongcun Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xuegang Luo
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Junqi Zhao
- School of Chemical and Biological Engineering, Qilu Institute of Technology, Jinan 250200, China
- Correspondence: (J.Z.); (Z.L.)
| | - Zhongyuan Li
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Correspondence: (J.Z.); (Z.L.)
| |
Collapse
|
106
|
Jeon SY, Jeon JH, Park JH, Lee J, Pang M, Choi MK, Song IS. Simultaneous Analysis of a Combination of Anti-Hypertensive Drugs, Fimasartan, Amlodipine, and Hydrochlorothiazide, in Rats Using LC-MS/MS and Subsequent Application to Pharmacokinetic Drug Interaction with Red Ginseng Extract. TOXICS 2022; 10:576. [PMID: 36287856 PMCID: PMC9610909 DOI: 10.3390/toxics10100576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
Fimasartan, amlodipine, and hydrochlorothiazide are commonly used in combination therapies as antihypertensive drugs. This study aimed to develop and validate an analytical method for fimasartan, its active and major metabolite fimasartan-amide, amlodipine, and hydrochlorothiazide in rat plasma using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The standard calibration curves for fimasartan (1−500 ng/mL), its active and major metabolite fimasartan-amide (0.3−100 ng/mL), amlodipine (0.5−200 ng/mL), and hydrochlorothiazide (5−5000 ng/mL) were linear with R2 > 0.9964, and the inter- and intra-day accuracy and precision and stability were within the acceptable criteria. Using this validated analytical method, the pharmacokinetic interaction of these triple combination drugs between single administration and concomitant administration of the triple combination was investigated; the results did not reveal a significant difference in any of the pharmacokinetic parameters. Based on these results, we investigated the effects of red ginseng extract (RGE) on the pharmacokinetics of fimasartan, fimasartan-amide, amlodipine, and hydrochlorothiazide after oral administration of the combination in rats. No significant difference was observed in the pharmacokinetic parameters of fimasartan, fimasartan-amide, amlodipine, and hydrochlorothiazide, except for the Tmax values of amlodipine. The delayed Tmax value of amlodipine was attributed to its decreased intestinal permeability after repeated RGE treatments. In conclusion, using a combination of antihypertensive drugs and simultaneous analytical methods, we established efficient drug interaction and toxicokinetic studies using a small number of animals.
Collapse
Affiliation(s)
- So-Yeon Jeon
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea
| | - Ji-Hyeon Jeon
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| | - Jin-Hyang Park
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| | - Jihoon Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| | - Minyeong Pang
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea
| | - Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
107
|
Design, synthesis, and biological evaluation of ocotillol derivatives fused with 2-aminothiazole via A-ring as modulators of P-glycoprotein-mediated multidrug resistance. Eur J Med Chem 2022; 243:114784. [PMID: 36167009 DOI: 10.1016/j.ejmech.2022.114784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/11/2022] [Accepted: 09/16/2022] [Indexed: 11/21/2022]
Abstract
Overexpression of P-glycoprotein (P-gp) plays a key role in the development of multidrug resistance (MDR), the major reason for the failure of chemotherapy in clinics. Ocotillol and its derivatives had been reported with good P-gp-mediated tumor MDR reversal activity in vitro. Herein, a series of ocotillol derivatives fused with 2-aminothiazole (2-AT) via A-ring were designed and synthesized to further improve the tumor MDR reversal potency. These compounds were evaluated for their MDR reversal activity against the KBV cells by MTT assay. Among them, the most promising derivative against P-gp-mediated MDR was compound 12 with 2-AT and glycine in the A-ring. Rhodamine123 (Rh123) accumulation assay, Western blot assay, and P-gp-Glo™ assay showed that compound 12 efficiently inhibited the efflux function of P-gp by stimulating P-gp ATPase rather than downregulating its expression. Moreover, compound 12 sensitized KBV cells to paclitaxel arrested cells in the G2/M phase and induced cell apoptosis. Importantly, compound 12 significantly inhibited the growth of KBV cell-derived xenograft tumors in nude mice by increasing the sensitivity of paclitaxel in vivo. Finally, the structure-activity relationships (SARs) of ocotillol derivatives were further investigated. In summary, compound 12 has the potential to overcome MDR in cancer caused by P-gp.
Collapse
|
108
|
Zhang J, Sayakoummane S, Kim SA, Lee JS, Choung ES, Kim ES, Lee SG, Yum J, Lee BH, Lee S, Kim JH, Cho JY. Hymenocallis littoralis ameliorates inflammatory responses in LPS-stimulated RAW264.7 cells and HCl/EtOH-induced gastric mucosal injury via targeting the MAPK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115400. [PMID: 35623503 DOI: 10.1016/j.jep.2022.115400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hymenocallis littoralis (Jacq.) Salisb. Also known as Pancratium littorale Jacq. And Hymenocallis panamensis Lindl., is a medicinal plant from the family Amarylideceae used for emetic and wound healing and has manifested anti-neoplastic, anti-oxidant, and anti-viral properties. AIM OF THE STUDY The aim of this paper is to investigate the anti-inflammatory potential and molecular mechanism of H. littoralis against lipopolysaccharide (LPS)-induced macrophages and in vivo HCl/EtOH-induced gastritis mucosal injury models. MATERIALS AND METHODS The production of pro-inflammatory cytokines and mediators was evaluated by Griess assay, RT-PCR, and real-time PCR. Moreover, the relevant proteins of mitogen-activated protein kinases (MAPKs) including ERK, JNK, p38, c-Jun, and c-Fos were detected using immunoblotting. RESULTS We demonstrated that H. littoralis prominently dampened production of nitric oxide (NO) in LPS-, poly I:C-, or pam3CSK-stimulated RAW264.7 cells; down-regulated the expression levels of interleukin 6 (IL-6) and inducible nitric oxide synthase; and markedly attenuated the luciferase activities of AP-1 reporter promoters. Moreover, H. littoralis administration prominently downregulated c-Fos and c-Jun phosphorylation as well as JNK1, ERK2, and MKK7 overexpression in HEK 293T cells. Furthermore, H. littoralis displayed anti-inflammatory effects in the HCl/EtOH-induced gastritis mice model. CONCLUSIONS Cumulatively, these results demonstrated that H. littoralis exerts eminently anti-inflammatory activities in LPS-stimulated RAW264.7 cells in vitro and in HCl/EtOH-induced gastritis mice models in vivo. These activities could be attributed to its modulatory effects on the MAPK signaling pathway.
Collapse
Affiliation(s)
- Jianmei Zhang
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Sousath Sayakoummane
- Department of Forestry, Ministry of Agriculture and Forestry, Vientiane Capital, P.O. Box 2932, Laos.
| | - Soo Ah Kim
- DanjoungBio Co. Ltd, Wonju, 26303, Republic of Korea.
| | - Jong Sub Lee
- DanjoungBio Co. Ltd, Wonju, 26303, Republic of Korea.
| | - Eui Su Choung
- DanjoungBio Co. Ltd, Wonju, 26303, Republic of Korea.
| | - Eun Sil Kim
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Seung-Gyu Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Jinwhoa Yum
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Byoung-Hee Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Sarah Lee
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, 22689, Republic of Korea.
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
109
|
Liu Y, Huang L, Kim MY, Cho JY. The Role of Thymoquinone in Inflammatory Response in Chronic Diseases. Int J Mol Sci 2022; 23:ijms231810246. [PMID: 36142148 PMCID: PMC9499585 DOI: 10.3390/ijms231810246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Anti-inflammatory therapies have been shown to be effective in the prevention of various cardiovascular diseases, tumors, and cancer complications. Thymoquinone (TQ), the main active constituent of Nigella sativa, has shown promising therapeutic properties in many in vivo and in vitro models. However, TQ has poor bioavailability and is hydrophobic, prohibiting clinical trials with TQ alone. Studies have explored the combination of TQ with biological nanomaterials to improve its bioavailability. The TQ nanoparticle formulation shows better bioavailability than free TQ, and these formulations are ready for clinical trials to determine their potential as therapeutic agents. In this paper, we review current knowledge about the interaction between TQ and the inflammatory response and summarize the research prospects in Korea and abroad. We discuss the different biological activities of TQ and various combination therapies of TQ and nanomaterials in clinical trials.
Collapse
Affiliation(s)
- Yan Liu
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Lei Huang
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea
| | - Mi-Yeon Kim
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.:+82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.:+82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
110
|
Jin GN, Lu JM, Lan HW, Lu YN, Shen XY, Xu X, Piao LX. Protective effect of ginsenoside Rh2 against Toxoplasma gondii infection-induced neuronal injury through binding TgCDPK1 and NLRP3 to inhibit microglial NLRP3 inflammasome signaling pathway. Int Immunopharmacol 2022; 112:109176. [PMID: 36067653 DOI: 10.1016/j.intimp.2022.109176] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is a neurotropic obligate intracellular parasite that can activate microglial and promote neuronal apoptosis, leading to central nervous system diseases. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling complex plays a key role in inducing neuroinflammation. Our previous studies have found that ginsenoside Rh2 (GRh2) inhibits T. gondii infection-induced microglial activation and neuroinflammation by downregulating the Toll-like receptor 4/nuclear factor-kappa B signaling pathway. However, whether GRh2 reduces T. gondii infection-induced neuronal injury through actions on microglial NLRP3 inflammasome signaling has not yet been clarified. METHODS In this study, we employed T. gondii RH strain to establish in vitro and in vivo infection models in BV2 microglia cell line and BALB/c mice. Molecular docking, localized surface plasmon resonance assay, quantitative competitive-PCR, ELISA, western blotting, flow cytometric analysis, and immunofluorescence were performed. RESULTS Our results showed that GRh2 alleviated neuropathological damage and neuronal apoptosis in cortical tissue of T. gondii-infected mice. GRh2 and CY-09 (an inhibitor of NLRP3) exhibited potent anti-T. gondii effects through binding T. gondii calcium-dependent protein kinase 1 (TgCDPK1). GRh2 decreased Iba-1 (a specific microglial marker) and NLRP3 inflammasome signaling pathway-related protein expression by binding NLRP3. Co-culture of microglia/primary cortical neurons revealed that T. gondii-induced microglial activation caused neuronal apoptosis, but GRh2 reduced this effect, consistent with the effects of CY-09. CONCLUSION Taken together, our results show that GRh2 has a protective effect against T. gondii infection-induced neuronal injury by binding TgCDPK1 and NLRP3 to inhibit NLRP3 inflammasome signaling pathway in microglia.
Collapse
Affiliation(s)
- Guang-Nan Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Jing-Mei Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Hui-Wen Lan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Yu-Nan Lu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Xin-Yu Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China
| | - Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China.
| | - Lian-Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin, China.
| |
Collapse
|
111
|
Hou J, Ma R, Zhu S, Wang Y. Revealing the Therapeutic Targets and Mechanism of Ginsenoside Rg1 for Liver Damage Related to Anti-Oxidative Stress Using Proteomic Analysis. Int J Mol Sci 2022; 23:ijms231710045. [PMID: 36077440 PMCID: PMC9455996 DOI: 10.3390/ijms231710045] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Ginsenoside Rg1 is an important active substance isolated from the root of ginseng. In previous studies, Rg1 has shown excellent therapeutic effects in antioxidant, anti-inflammatory, and metabolic modulation. However, the therapeutic targets of Rg1 are still unknown. In this study, we investigated the therapeutic effects of Rg1 on oxidative stress-related liver damage. The oxidative stress damage model was achieved by intraperitoneal injection of D-galactose (D-gal) for 42 consecutive days in C57BL/6J mice. Rg1 treatment started on Day 16. Body weight, liver weight, degree of hepatic oxidative stress damage, serum lipid levels, and hepatic lipid and glucose metabolism were measured. Proteomics analysis was used to measure liver protein expression. The differential expression proteins were analyzed with bioinformatics. The results showed that Rg1 treatment attenuated liver damage from oxidative stress, reduced hepatic fat accumulation, promoted hepatic glycogen synthesis, and attenuated peripheral blood low-density lipoprotein (LDL), cholesterol (CHO), and triglycerides (TG) levels. Proteomic analysis suggested that Rg1 may regulate hepatocyte metabolism through ECM-Receptor, the PI3K-AKT pathway. The epidermal growth factor receptor (EGFR) and activator of transcription 1 (STAT1) may be the key protein. In conclusion, this study provides an experimental basis for further clarifying the specific mechanism of Rg1 in the treatment of oxidative stress damage-related liver disease.
Collapse
Affiliation(s)
- Jiying Hou
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
| | - Ruoxiang Ma
- Faculty of Basic Medical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Shisheng Zhu
- Faculty of Basic Medical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
- Correspondence: (S.Z.); (Y.W.)
| | - Yaping Wang
- Laboratory of Stem Cells and Tissue Engineering, Department of Histology and Embryology, Chongqing Medical University, Chongqing 400016, China
- Correspondence: (S.Z.); (Y.W.)
| |
Collapse
|
112
|
Lu J, Yu Y, Wang XJ, Chai RP, Lyu XK, Deng MH, Hu MG, Qi Y, Chen X. Mechanism of Shengmai Injection on Anti-Sepsis and Protective Activities of Intestinal Mucosal Barrier in Mice. Chin J Integr Med 2022; 28:817-822. [PMID: 34241804 DOI: 10.1007/s11655-021-3292-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To study the mechanism of Shengmai Injection (SMI, ) on anti-sepsis and protective activities of intestinal mucosal barrier. METHODS The contents of 11 active components of SMI including ginsenoside Rb1, Rb2, Rb3, Rd, Re, Rf, Rg1, Rg2, ophioposide D, schisandrol A and schisantherin A were determined using ultra-performance liquid chromatography. Fifty mice were randomly divided into the blank, the model, the low-, medium- and high-dose SMI groups (0.375, 0.75, 1.5 mL/kg, respectively) by random number table, 10 mice in each group. In SMI group, SMI was administrated to mice daily via tail vein injection for 3 consecutive days, while the mice in the blank and model groups were given 0.1 mL of normal saline. One hour after the last SMI administration, except the blank group, the mice in other groups were intraperitoneally injected with lipopolysaccharide (LPS) saline solution (2 mL/kg) at a dosage of 5 mL/kg for development of endotoxemia mice model. The mice in the blank group were given the same volume of normal saline. Inflammatory factors including interferon-γ (INF-γ), tumor necrosis factor-α (TNF-α), interleukin (IL)-2 and IL-10 were measured by flow cytometry. Myosin light-chain kinase (MLCK), nuclear factor κB (NF-κB) levels, and change of Occludin proteins in jejunum samples were analyzed by Western blot. RESULTS The decreasing trends of INF-γ, TNF-α and IL-2 were found in serum of SMI treatment groups. In SMI-treated mice, the content of Occludin increased and MLCK protein decreased compared with the model group (P<0.05 or P<0.01). The content of cellular and nuclear NF-κB did not change significantly (P>0.05). CONCLUSION SMI may exert its anti-sepsis activity mainly through NF-κB-pro-inflammatory factor-MLCK-TJ cascade.
Collapse
Affiliation(s)
- Juan Lu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Yue Yu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Xiao-Jing Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Rui-Ping Chai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Xin-Kai Lyu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Ming-Hui Deng
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Mei-Geng Hu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China
| | - Xi Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
113
|
Lee SG, Cho KH, Nguyen TTL, Vo DK, Chae YJ, Maeng HJ. Inhibitory effect of 20(S)-protopanaxadiol on cytochrome P450: Potential of its pharmacokinetic interactions in vivo. Biomed Pharmacother 2022; 153:113514. [DOI: 10.1016/j.biopha.2022.113514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/24/2022] [Accepted: 08/02/2022] [Indexed: 12/28/2022] Open
|
114
|
Liu S, Ju AQ, Duan AY, Zhan CL, Gao LP, Ma X, Yang SB. Presence of Ginsenoside Re during in vitro maturation protects porcine oocytes from heat stress. Reprod Domest Anim 2022; 57:1572-1583. [PMID: 36001037 DOI: 10.1111/rda.14234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/10/2022] [Accepted: 08/20/2022] [Indexed: 11/30/2022]
Abstract
Heat stress (HS) affects the development of porcine gametes and embryos negatively, induces the decrease of reproductive ability significantly, threatens global pig production. Ginsenoside Re (GRe), is a main bioactive component of ginseng, shows very specific anti-apoptotic, antioxidant and anti-inflammatory activities. To investigate the alleviating effect of GRe on the in vitro maturation of porcine oocyte under the HS, the polar body extrusion rate, intracellular levels of reactive oxygen species (ROS) and glutathione (GSH), ATP content, mitochondrial membrane potential (MMP) were assessed. For the current study, porcine cumulus-oocyte complexes (COCs) randomly divided into four groups: the control, GRe, HS and HS+GRe group. The results showed that HS inhibited the cumulus cell expansion and polar body extrusion rate, the levels of GSH and MMP, the ATP content, the gene expression of Nrf2 of porcine oocytes and the parthenogenetic activation (PA) embryo development competence, but GRe treatment could partly neutralize these adverse effects. Moreover, HS increased the ROS formation and percentage of apoptosis, the gene expression of HSP90, CASP3 and CytoC of porcine oocytes, but GRe could weaken the effect on Cyto C and BAX expression induced by HS. Taken together, these results showed that the presence of GRe during in vitro maturation protects porcine oocytes from HS. These findings lay a foundation for GRe may be used as a potential protective drug to protect porcine oocytes against HS damage.
Collapse
Affiliation(s)
- Shuang Liu
- College of Animal Science and Technology, Jilin Agricultural University, Jilin, China
| | - An-Qi Ju
- College of Animal Science and Technology, Jilin Agricultural University, Jilin, China
| | - Ao-Yi Duan
- College of Animal Science and Technology, Jilin Agricultural University, Jilin, China
| | - Cheng-Lin Zhan
- College of Animal Science and Technology, Jilin Agricultural University, Jilin, China
| | - Le-Peng Gao
- College of Animal Science and Technology, Jilin Agricultural University, Jilin, China
| | - Xin Ma
- College of Animal Science and Technology, Jilin Agricultural University, Jilin, China
| | - Shu-Bao Yang
- Center for Reproductive Medicine, Jilin Medical University, Jilin, China
| |
Collapse
|
115
|
Hepatoprotective Mechanism of Ginsenoside Rg1 against Alcoholic Liver Damage Based on Gut Microbiota and Network Pharmacology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5025237. [PMID: 36052161 PMCID: PMC9427247 DOI: 10.1155/2022/5025237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
Alcoholic liver disease (ALD) is a major public health problem worldwide, which needs to be effective prevention. Ginsenoside Rg1 (GRg1), a bioactive ingredient extracted from ginseng, has benefit effects on health. In this study, 11 potential targets of GRg1 against ALD were firstly obtained by network pharmacology. KEGG pathway enrichment showed that GRg1-target-ALD was closely related to Toll-like receptor (TLR) and nuclear factor-kappa B (NF-κB) signaling pathways. In addition, GRg1 decreased antioxidant levels and increased oxidative levels in alcohol-treated mice, which alleviated oxidative stress-induced hepatic damage. GRg1 enhanced intestinal barrier function via upregulating the levels of tight junction protein and immunoglobulin A. GRg1 also reduced alcohol-induced inflammation by suppressing TLR4/NF-κB pathway, which was consistent with the prediction of network targets. Moreover, GRg1 altered GM population, and Verrucomicrobia, Bacteroidetes, Akkermansia, Bacteroides, Lachnospiraceae_NK4A136_group, and Alloprevotella played positive association with intestinal barrier indicators and negative correlation with hepatic inflammation biomarkers. The results suggest that GRg1 administration might be a promising strategy for protection of alcohol-induced liver damage.
Collapse
|
116
|
New Therapeutic Approaches to and Mechanisms of Ginsenoside Rg1 against Neurological Diseases. Cells 2022; 11:cells11162529. [PMID: 36010610 PMCID: PMC9406801 DOI: 10.3390/cells11162529] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Neurological diseases, including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), stroke, cerebral infarction, ischemia-reperfusion injury, depression and, stress, have high incidence and morbidity and often lead to disability. However, there is no particularly effective medication against them. Therefore, finding drugs with a suitable efficacy, low toxicity and manageable effects to improve the quality of life of patients is an urgent problem. Ginsenoside Rg1 (Rg1) is the main active component of ginseng and has a variety of pharmacological effects. In this review, we focused on the therapeutic potential of Rg1 for improving neurological diseases. We introduce the mechanisms of Ginsenoside Rg1 in neurological diseases, including apoptosis, neuroinflammation, the microRNA (miRNA) family, the mitogen-activated protein kinase (MAPK) family, oxidative stress, nuclear factor-κB (NF-κB), and learning and memory of Rg1 in neurological diseases. In addition, Rg1 can also improve neurological diseases through the interaction of different signal pathways. The purpose of this review is to explore more in-depth ideas for the clinical treatment of neurological diseases (including PD, AD, HD, stroke, cerebral infarction, ischemia–reperfusion injury, depression, and stress). Therefore, Rg1 is expected to become a new therapeutic method for the clinical treatment of neurological diseases.
Collapse
|
117
|
Han L, Li T, Miao D, Lee J, Xiao S, Piao H, Zhao Y. Design, Synthesis and Cytotoxicity Evaluation of Novel Indole Derivatives of Panaxadiol. Chem Biodivers 2022; 19:e202200372. [PMID: 35938749 DOI: 10.1002/cbdv.202200372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/18/2022] [Indexed: 11/05/2022]
Abstract
Based on the well-known cytotoxicity of indole compounds, we used the 'Fisher indole synthesis' method to introduce appropriately substituted indole rings into panaxadiol (PD), generating eighteen novel Panaxadiol indole derivatives. Six human cancer cell lines (A549, HepG-2, HCT-116, SGC-7901, MDA-MB-231, PC-3 cells) and one normal ovarian cell lines (IOSE144) were designed to evaluate the anti-proliferative activity of the PD derivatives. The results showed that the majority of PD derivatives showed enhanced anti-proliferative activity, when compared with PD, with P-Methylindolo-PD exhibiting the highest cytotoxicity. In A549 cells, IC50 value was 5.01±0.87 μM, which is roughly 12 times higher than the activity of PD and 5 times that of 5-FU. Moreover, cell morphology analysis and Annexin V-FITC/PI assays exhibited that P-Methylindolo-PD could induce A549 cell apoptosis (55.7 % of apoptotic cells at 20 μM). Moreover, molecular docking experiments were performed to explore the molecular mechanism underlining the binding of P-Methylindolo-PD to the active site of EGFR. The results support that P-Methylindolo-PD might be a promising lead compound for further studies.
Collapse
Affiliation(s)
- Linlin Han
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| | - Tao Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| | - Dongyu Miao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| | - Jungjoon Lee
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| | - Shengnan Xiao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| | - Huri Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| | - Yuqing Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, P. R. China
| |
Collapse
|
118
|
Tu Y, Li L, Fan W, Liu L, Wang Z, Yang L. Development of Green and Efficient Extraction of Bioactive Ginsenosides from Panax ginseng with Deep Eutectic Solvents. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144339. [PMID: 35889211 PMCID: PMC9315546 DOI: 10.3390/molecules27144339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/28/2022]
Abstract
The extraction of active constituents from natural sources in a green and efficient manner is considered an important field in the pharmaceutical industry. In recent years, deep eutectic solvents (DESs), a new type of green solvent, have attracted increasing attention. Therefore, we aimed to establish a green and high-efficiency extraction method for ginsenosides based on DESs. This study takes Panax ginseng as a model sample. Eighteen different DESs were produced to extract polar ginsenosides. Ultrasound-assisted extraction (UAE) was applied for simplicity and efficiency. A binary DES synthesized using choline chloride and urea at a proportion of 1:2 prepared by a heating stirring method is proven to be more effective than other solvents, such as the widely used 70% ethanol for the extraction of ginsenosides. Three variables that might affect the extraction, including the DES content in the extraction solvent, liquid/solid ratio, and ultrasound extraction time, were evaluated for optimization. The optimum extraction conditions for ginsenosides were determined as follows: DES water content of 20 wt%, liquid/solid ratio of 15 mL g−1, and an ultrasonic extraction time of 15 min. The extraction yield for the optimized method is found to be 31% higher than that for 70% ethanol, which achieves efficient extraction. This study shows that DESs are available to extract ginsenosides for use in traditional Chinese medicine. The discovery also contributes to further research into the green extraction of ginsenosides.
Collapse
Affiliation(s)
- Yujia Tu
- The MOE Key Laboratory of Standardization of Chinese Medicines, The SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.T.); (W.F.); (L.L.); (Z.W.)
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicines, Shanghai 201203, China
| | - Linnan Li
- The MOE Key Laboratory of Standardization of Chinese Medicines, The SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.T.); (W.F.); (L.L.); (Z.W.)
- Correspondence: (L.L.); (L.Y.); Tel.: +86-021-51322506 (L.L.); +86-021-51322506 (L.Y.)
| | - Wenxiang Fan
- The MOE Key Laboratory of Standardization of Chinese Medicines, The SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.T.); (W.F.); (L.L.); (Z.W.)
| | - Longchan Liu
- The MOE Key Laboratory of Standardization of Chinese Medicines, The SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.T.); (W.F.); (L.L.); (Z.W.)
| | - Zhengtao Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, The SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.T.); (W.F.); (L.L.); (Z.W.)
| | - Li Yang
- The MOE Key Laboratory of Standardization of Chinese Medicines, The SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicine, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.T.); (W.F.); (L.L.); (Z.W.)
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicines, Shanghai 201203, China
- Correspondence: (L.L.); (L.Y.); Tel.: +86-021-51322506 (L.L.); +86-021-51322506 (L.Y.)
| |
Collapse
|
119
|
Kim J, Phung HM, Lee S, Kim KT, Son TK, Kang KS, Lee S. Anti-skin-aging effects of tissue-cultured mountain-grown ginseng and quantitative HPLC/ELSD analysis of major ginsenosides. J Nat Med 2022; 76:811-820. [PMID: 35748997 DOI: 10.1007/s11418-022-01633-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/28/2022] [Indexed: 11/24/2022]
Abstract
Mountain-grown ginseng has free radical scavenging activity and suppresses inflammation. We evaluated the anti-skin-aging effects of tissue-cultured mountain-grown ginseng (TG) and its major ginsenosides. The effect of three extracts of TG and ginsenosides Rg1 (1), Rf (2), Rb1 (3), Re (4), and Rd (5) on the secretion of matrix metalloproteinase-1 (MMP-1) and collagen type I alpha 1 (COLIA1) was compared with that of tumor necrosis factor-alpha (TNF-α) stimulation of human dermal fibroblasts (HDFs), as determined via enzyme-linked immunosorbent assay. An analytical high-performance liquid chromatographic method with evaporative light-scattering detection (HPLC/ELSD) was developed for the simultaneous determination of the major ginsenosides in TG obtained via supercritical fluid CO2 or ethanol extraction. TG residues obtained via supercritical fluid CO2 extraction (TG1) and TG not subject to extraction (TG3) suppressed MMP-1 secretion in TNF-α-stimulated HDFs. Major ginsenoside content was higher in the TG1 than in residues extracted with ethanol (TG2) and TG3; ginsenoside Rg1 (1) content was the highest among all TG residues. Among them, ginsenosides Rg1 (1) and Re (4) suppressed MMP-1 in TNF-α-stimulated HDFs, whereas ginsenosides Rb1 (3) and Rd (5) increased COLIA1. In conclusion, TG and its active ginsenosides may have anti-skin-aging effects. Ginsenoside Rg1 (1) may also be beneficial in ameliorating skin damage. HPLC/ELSD can identify major ginsenosides and supercritical fluid CO2 extraction can be applied during health supplement or drug development.
Collapse
Affiliation(s)
- Juree Kim
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, Korea
| | - Hung Manh Phung
- Department of Preventive Medicine, Gachon University, Seongnam, 13120, Korea
| | - Sullim Lee
- Department of Life Science, Gachon University, Seongnam, 13120, Korea
| | | | - Tae Kwon Son
- Department of Farm Management, Kyungpook National University, Daegu, 41566, Korea.,Apple Bio Co. Ltd, Daegu, 41566, Korea
| | - Ki Sung Kang
- Department of Preventive Medicine, Gachon University, Seongnam, 13120, Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, 17546, Korea. .,BET Research Institute, Chung-Ang University, Anseong, 17546, Korea. .,Natural Product Institute of Science and Technology, Anseong, 17546, Korea.
| |
Collapse
|
120
|
Ye XW, Wang HL, Cheng SQ, Xia LJ, Xu XF, Li XR. Network Pharmacology-Based Strategy to Investigate the Pharmacologic Mechanisms of Coptidis Rhizoma for the Treatment of Alzheimer's Disease. Front Aging Neurosci 2022; 14:890046. [PMID: 35795239 PMCID: PMC9252849 DOI: 10.3389/fnagi.2022.890046] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
BackgroundAlzheimer's disease (AD) is becoming a more prevalent public health issue in today's culture. The experimental study of Coptidis Rhizoma (CR) and its chemical components in AD treatment has been widely reported, but the principle of multi-level and multi-mechanism treatment of AD urgently needs to be clarified.ObjectiveThis study focuses on network pharmacology to clarify the mechanism of CR's multi-target impact on Alzheimer's disease.MethodsThe Phytochemical-compounds of CR have been accessed from the Traditional Chinese Medicine Database and Analysis Platform (TCMSP) and Symmap database or HPLC determination. The values of Oral Bioavailability (OB) ≥ 30% and Drug Like (DL) ≥ 0.18 or blood ingredient were used to screen the active components of CR; the interactive network of targets and compounds were constructed by STRING and Cytoscape platform, and the network was analyzed by Molecular Complex Detection (MCODE); Gene Ontology (GO) function, Kyoto Encyclopedia of Genes and Genomes Pathway (KEGG) and metabolic pathway enrichment of targets were carried out with Metascape, the Database for Annotation, Visualization and Integrated Discovery (DAVID) and MetaboAnalyst platform; Based on CytoHubba, the potential efficient targets were screened by Maximal Clique Centrality (MCC) and Degree, the correlation between potential efficient targets and amyloid β-protein (Aβ), Tau pathology was analyzed by Alzdata database, and the genes related to aging were analyzed by Aging Altas database, and finally, the core targets were obtained; the binding ability between ingredients and core targets evaluated by molecular docking, and the clinical significance of core targets was assessed with Gene Expression Omnibus (GEO) database.Results19 active components correspond to 267 therapeutic targets for AD, of which 69 is potentially effective; in module analysis, RELA, TRAF2, STAT3, and so on are the critical targets of each module; among the six core targets, RELA, MAPK8, STAT3, and TGFB1 have clinical therapeutic significance; GO function, including 3050 biological processes (BP), 257 molecular functions (MF), 184 cellular components (CC), whose functions are mainly related to antioxidation, regulation of apoptosis and cell composition; the HIF-1 signaling pathway, glutathione metabolism is the most significant result of 134 KEGG signal pathways and four metabolic pathways, respectively; most of the active components have an excellent affinity in docking with critical targets.ConclusionThe pharmacological target prediction of CR based on molecular network pharmacology paves the way for a multi-level networking strategy. The study of CR in AD treatment shows a bright prospect for curing neurodegenerative diseases.
Collapse
Affiliation(s)
- Xian-wen Ye
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hai-li Wang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Shui-qing Cheng
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Liang-jing Xia
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xin-fang Xu
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Xin-fang Xu
| | - Xiang-ri Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Xiang-ri Li
| |
Collapse
|
121
|
The Efficacy of Ginseng (Panax) on Human Prediabetes and Type 2 Diabetes Mellitus: A Systematic Review and Meta-Analysis. Nutrients 2022; 14:nu14122401. [PMID: 35745129 PMCID: PMC9227417 DOI: 10.3390/nu14122401] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Results from different clinical trials on the effects of ginseng on prediabetes and type 2 diabetes (T2DM) are still inconsistent. To fill this knowledge gap, we investigated the overall effects of ginseng supplementation on improving cardiometabolic biomarkers among these patients. A systematic literature search was conducted on PubMed/MEDLINE, Scopus, Web of Science, and Cochrane library. A random-effect model was applied to estimate the weighted mean difference and 95% CI for each outcome. Overall, 20 eligible RCTs were included. Meta-analyses revealed that ginseng supplementation significantly reduced serum concentration of FPG, TC, IL-6, and HOMA-IR values. It also increased HR and TNF-α levels. Ginseng supplementation changed HOMA-IR and HDL-C significantly based on dose and changed HOMA-IR and LDL-C significantly based on study duration in a non-linear fashion. Furthermore, meta-regression analyses indicated a linear relationship between ginseng dose and absolute changes in HDL-C. Moreover, subgroup analyses showed that ginseng supplementation changed TC and LDL-C when the supplementation dose was ≥2 g/day. Our findings suggest that ginseng supplementation may be an effective strategy for improving cardiometabolic profiles in individuals with prediabetes and T2DM.
Collapse
|
122
|
Screening and Isolation of Potential Anti-Inflammatory Compounds from Saxifraga atrata via Affinity Ultrafiltration-HPLC and Multi-Target Molecular Docking Analyses. Nutrients 2022; 14:nu14122405. [PMID: 35745138 PMCID: PMC9230087 DOI: 10.3390/nu14122405] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
In this study, a 100 g sample of Saxifraga atrata was processed to separate 1.3 g of 11-O-(4′-O-methylgalloyl)-bergenin (Fr1) after 1 cycle of MCI GEL® CHP20P medium pressure liquid chromatography using methanol/water. Subsequently, COX-2 affinity ultrafiltration coupled with reversed-phase liquid chromatography was successfully used to screen for potential COX-2 ligands in this target fraction (Fr1). After 20 reversed-phase liquid chromatography runs, 74.1 mg of >99% pure 11-O-(4′-O-methylgalloyl)-bergenin (Fr11) was obtained. In addition, the anti-inflammatory activity of 11-O-(4′-O-methylgalloyl)-bergenin was further validated through molecular docking analyses which suggested it was capable of binding strongly to ALOX15, iNOS, ERBB2, SELE, and NF-κB. As such, the AA metabolism, MAPK, and NF-κB signaling pathways were hypothesized to be the main pathways through which 11-O-(4′-O-methylgalloyl)-bergenin regulates inflammatory responses, potentially functioning by reducing pro-inflammatory cytokine production, blocking pro-inflammatory factor binding to cognate receptors and inhibiting the expression of key proteins. In summary, affinity ultrafiltration-HPLC coupling technology can rapidly screen for multi-target bioactive components and when combined with molecular docking analyses, this approach can further elucidate the pharmacological mechanisms of action for these compounds, providing valuable information to guide the further development of new multi-target drugs derived from natural products.
Collapse
|
123
|
El-Banna MA, Hendawy OM, El-Nekeety AA, Abdel-Wahhab MA. Efficacy of ginsenoside Rg3 nanoparticles against Ehrlich solid tumor growth in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:43814-43825. [PMID: 35118592 DOI: 10.1007/s11356-022-19019-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Solid tumors are fairly common and face many clinical difficulties since they are hardly surgically resectable and broadly do not respond to radiation and chemotherapy. The current study aimed to fabricate ginsenoside Rg3 nanoparticles (Rg3-NPs) and evaluate their antitumor effect against Ehrlich solid tumors (EST) in mice. Rg3-NPs were fabricated using whey protein isolates (WPI), maltodextrin (MD), and gum Arabic (GA). EST was developed by the injection of mice with Ehrlich ascites cells (2.5 × 106). The mice were divided into a control group, EST group, and the EST groups that were treated orally 2 weeks for with normal Rg3 (3 mg/kg b.w.), Rg3-NPs at a low dose (3 mg/kg b.w.), and Rg3-NPs at a high dose (6 mg/kg b.w.). Serum and solid tumors were collected for different assays. The results revealed that synthesized Rg3-NPs showed a spherical shape with an average particle size of 20 nm and zeta potential of -5.58 mV. The in vivo study revealed that EST mice showed a significant increase in AFP, Casp3, TNF-α, MMP-9, VEGF, MDA, and DNA damage accompanied by a significant decrease in SOD and GPx. Treatment with Rg3 or Rg3-NPs decreased the tumor weight and size and induced a significant improvement in all the biochemical parameters. Rg3-NPs were more effective than Rg3, and the improvement was dose-dependent. It could be concluded that fabrication of Rg3-NPs enhanced the protective effect against EST development which may be due to the synergistic effect of Rg3 and MD, GA, and WPI.
Collapse
Affiliation(s)
- Mona A El-Banna
- Medical Biochemistry Department, National Research Centre, Dokki, Cairo, Egypt
| | - Omnia M Hendawy
- Clinical Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Aziza A El-Nekeety
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt
| | - Mosaad A Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Centre, Dokki, Cairo, Egypt.
| |
Collapse
|
124
|
Tran THM, Puja AM, Kim H, Kim YJ. Nanoemulsions prepared from mountain ginseng-mediated gold nanoparticles and silydianin increase the anti-inflammatory effects by regulating NF-κB and MAPK signaling pathways. BIOMATERIALS ADVANCES 2022; 137:212814. [PMID: 35929253 DOI: 10.1016/j.bioadv.2022.212814] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/18/2022] [Accepted: 04/16/2022] [Indexed: 06/15/2023]
Abstract
In order to increase the bioavailability of mountain ginseng (MG), gold nanoparticles (MG-AuNPs) were biologically synthesized from MG extract, and an oil-in-water (O/W) nanoemulsion (SMG-AuNEs) was prepared from MG-AuNPs and a phytochemical silydianin. The physical stability of SMG-AuNEs were monitored and optimized in terms of particle size, pH value, zeta potential, and polydispersity index. The chemicostructural properties of the prepared MG-AuNPs and SMG-AuNEs were characterized using various spectrometric and microscopic analyses, such as EDX spectroscopy, FT-IR spectroscopy, and TEM. The effect of both nanomaterial samples on the anti-inflammatory activity and their underlying mechanism was compared in LPS-stimulated RAW 264.7 cells. SMG-AuNEs did not show toxic effects against RAW 264.7 macrophages, HaCaT keratinocytes, and normal dermal fibroblasts. SMG-AuNEs exhibited significantly higher inhibition of pro-inflammatory genes and proteins, including IL-1β, IL-6, and TNF-α, compared with those of MG-AuNPs and silydianin. Western blotting analysis revealed that the MAPK and NF-κB signalings were highly inhibited by SMG-AuNEs treatment. Hence, this study shows that nano-emulsification of gold nanoparticles prepared from MG is a useful method for augmenting the anti-inflammatory potential of MG. This study may serve as a foundation for using MG as a functional ingredient in anti-inflammatory agents. Our results may implicate the use of nanoemulsions to develop new anti-inflammatory products using MG.
Collapse
Affiliation(s)
- Thi Hoa My Tran
- Department of Oriental Medicine Biotechnology, Kyung Hee University, Yongin 17104, Gyeonggi, Republic of Korea
| | - Aditi Mitra Puja
- Department of Oriental Medicine Biotechnology, Kyung Hee University, Yongin 17104, Gyeonggi, Republic of Korea
| | - Hoon Kim
- Department of Oriental Medicine Biotechnology, Kyung Hee University, Yongin 17104, Gyeonggi, Republic of Korea.
| | - Yeon-Ju Kim
- Department of Oriental Medicine Biotechnology, Kyung Hee University, Yongin 17104, Gyeonggi, Republic of Korea.
| |
Collapse
|
125
|
Kang J, Zhou Y, Zhu C, Ren T, Zhang Y, Xiao L, Fang B. Ginsenoside Rg1 Mitigates Porcine Intestinal Tight Junction Disruptions Induced by LPS through the p38 MAPK/NLRP3 Inflammasome Pathway. TOXICS 2022; 10:toxics10060285. [PMID: 35736894 PMCID: PMC9228030 DOI: 10.3390/toxics10060285] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
Abstract
Inflammation leads to porcine tight junction disruption of small intestinal epithelial cells, resulting in intestinal dysfunction. Herein, we established lipopolysaccharide (LPS)-induced in-vivo and in-vitro inflammatory models. The results revealed that LPS induced tight junction disruption in IPEC-J2 cells by downregulating tight-junction-related protein zonula occludens-1 (ZO-1), occludin and claudin-1 expression, while ginsenoside Rg1 rescued such inhibition and abrogated the upregulated expression of phosphorylation p38 MAPK. The p38 MAPK inhibitor (SB203580) showed a similar effect with Rg1 and attenuated the LPS-induced inhibition of ZO-1, occludin and claudin-1 expression, which is consistent with the reduced expression of NLRP3 inflammasome and IL-1β. Furthermore, the specific inhibitors of NLRP3 and IL-1β result in increased expression of tight-junction-related protein, demonstrating that p38 MAPK signaling was associated with Rg1 suppression of tight junction disruption. Besides, LPS treatment decreased the expression of ZO-1, occludin and claudin-1 through p38 MAPK signaling, and caused abnormal morphological changes in murine ileum. Meanwhile, Rg1 attenuated the decreased expression of ZO-1, occludin and claudin-1 and partially alleviated LPS-induced morphological changes in murine ileum. In summary, these findings characterized a novel mechanism by which Rg1 alleviates LPS-induced intestinal tight junction disruption by inhibiting the p38 MAPK-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Jian Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Yanhong Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Chunyang Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Tian Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 100096, China
- Correspondence: (L.X.); (B.F.)
| | - Binghu Fang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510630, China; (J.K.); (Y.Z.); (C.Z.); (T.R.)
- Correspondence: (L.X.); (B.F.)
| |
Collapse
|
126
|
Hossain MA, Kim JH. Possibility as role of ginseng and ginsenosides on inhibiting the heart disease of COVID-19: A systematic review. J Ginseng Res 2022; 46:321-330. [PMID: 35068945 PMCID: PMC8767974 DOI: 10.1016/j.jgr.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/25/2022] Open
Abstract
Coronavirus has been spreading rapidly around the world since it broke out in China in 2019. Respiratory diseases caused by coronavirus infection cause various diseases ranging from asymptomatic subclinical infections to severe pneumonia and cardiovascular complications, leading to death. In this regard, natural products are being studied to prevent various diseases caused by COVID-19. In current review, we would like to present mechanisms related to the inhibition of heart disease in ginseng and ginsenoside against SARS-CoV-2. In many previous studies, ginseng and ginsenoside are known to have antioxidant, blood flow improvement, improvement of vascular and heart function, blood pressure control, suppression of myocardial infarction and heart failure, and antiarrhythmia. Therefore, ginseng and ginsenoside have a possibility to suppress cardiovascular complications caused by COVID-19. Many of research provide evidence for ginseng and ginsenoside as treatments for the risk of cardiovascular complications. However, in this review, more specific contents on the proposition of the efficacy of ginseng and ginsenoside for COVID-19 should be presented. Therefore, we hope that researches to reduce cardiovascular complications of ginseng and ginsenoside for COVID-19 should be presented to reduce mortality for COVID-19.
Collapse
Affiliation(s)
- Mohammad Amjad Hossain
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| |
Collapse
|
127
|
Wang DS, Wang JM, Zhang FR, Lei FJ, Wen X, Song J, Sun GZ, Liu Z. Ameliorative Effects of Malonyl Ginsenoside from Panax ginseng on Glucose-Lipid Metabolism and Insulin Resistance via IRS1/PI3K/Akt and AMPK Signaling Pathways in Type 2 Diabetic Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:863-882. [PMID: 35282802 DOI: 10.1142/s0192415x22500367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our previous study has revealed that malonyl-ginsenosides from Panax ginseng (PG-MGR) play a crucial role in the treatment of T2DM. However, its potential mechanism was still unclear. In this study, we investigated the anti-diabetic mechanisms of action of PG-MGR in high fat diet-fed (HFD) and streptozotocin-induced diabetic mice and determined the main constituents of PG-MGR responsible for its anti-diabetic effects. Our results showed that 16 malonyl ginsenosides were identified in PG-MGR by HPLC-ESI-MS/MS. PG-MGR treatment significantly reduced fasting blood glucose (FBG), triglyceride (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels and improved insulin resistance and glucose tolerance. Simultaneously, PG-MGR treatment improved liver injury by decreasing aspartate aminotransferase (AST) and alanine aminotransferase (ALT) expression. Furthermore, Western blot analysis demonstrated that the protein expression levels of p-PI3K/PI3K, p-AKT/AKT, p-AMPK/AMPK, p-ACC/ACC and GLUT4 in liver and skeletal muscle were significantly up-regulated after PG-MGR treatment, and the protein expression levels of p-IRS-1/IRS-1, Fas and SREBP-1c were significantly reduced. These findings revealed that PG-MGR has the potential to improve glucose and lipid metabolism and insulin resistance by activating the IRS-1/PI3K/AKT and AMPK signal pathways.
Collapse
Affiliation(s)
- Dong-Sheng Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jia-Mei Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Fu-Rui Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Feng-Jie Lei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Xin Wen
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jia Song
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Guang-Zhi Sun
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| |
Collapse
|
128
|
Shin JE, Jeon SH, Lee SJ, Choung SY. The Administration of Panax Ginseng Berry Extract Attenuates High-Fat-Diet-Induced Sarcopenic Obesity in C57BL/6 Mice. Nutrients 2022; 14:nu14091747. [PMID: 35565712 PMCID: PMC9099595 DOI: 10.3390/nu14091747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 01/05/2023] Open
Abstract
Sarcopenia and obesity are serious health problems that are highly related to several metabolic diseases. Sarcopenic obesity, a combined state of sarcopenia and obesity, results in higher risks of metabolic diseases and even mortality than sarcopenia or obesity alone. Therefore, the development of therapeutic agents for sarcopenic obesity is crucial. C57BL/6 mice were fed with a high-fat diet (HFD) for 9 weeks. Then, mice were administered with Panax ginseng berry extract (GBE) for an additional 4 weeks, with continuous HFD intake. GBE significantly decreased the food efficiency ratio, serum lipid and insulin levels, adipose tissue weights, and adipocyte size. It significantly increased the grip strength, muscle masses, and myofiber cross-sectional area. It deactivated the protein kinase C (PKC) theta and zeta, resulting in activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, which is known to regulate muscle synthesis and degradation. Furthermore, it inhibited the production of inflammatory cytokines in the muscle tissue. GBE attenuated both obesity and sarcopenia. Thus, GBE is a potential agent to prevent or treat sarcopenic obesity.
Collapse
Affiliation(s)
- Ji-Eun Shin
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - So-Hyun Jeon
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | | | - Se-Young Choung
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea;
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-9198; Fax: +82-2-961-0372
| |
Collapse
|
129
|
Bi S, Qu Y, Shao J, Zhang J, Li W, Zhang L, Ni J, Cao L. Ginsenoside Rg3 Ameliorates Stress of Broiler Chicks Induced by Escherichia coli Lipopolysaccharide. Front Vet Sci 2022; 9:878018. [PMID: 35464384 PMCID: PMC9024239 DOI: 10.3389/fvets.2022.878018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 11/30/2022] Open
Abstract
In broiler chicks, Escherichia coli lipopolysaccharide is a prominent cause for inflammatory damage and loss of immune homeostasis in broiler chicks. Ginsenosides have been shown to have anti-inflammatory and antioxidant effects. However, it has not been demonstrated that ginsenosides protect broiler chicks against stress induced by Escherichia coli lipopolysaccharide challenge. The aim of this is to investigate the protective effect of ginsenosides Rg1, Re, and Rg3 on Escherichia coli lipopolysaccharide-induced stress. Our results showed that Rg3 ameliorated growth inhibition and fever, as well as decreased the production of stress-related hormones in broilers with stress. The protective effect of Rg3 on the stressed chicks may be largely mediated by regulating inflammatory response and oxidative damage. Moreover, real-time quantitative-polymerase chain reaction (RT-qPCR) results demonstrated that Rg3 upregulated mRNA expression of mTOR, HO-1, and SOD-1. These results suggested that ginsenoside Rg3 and ginsenoside products contains Rg3 deserve further study for the control of immunological stress and inflammation in broiler chicks.
Collapse
Affiliation(s)
- Shicheng Bi
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
- *Correspondence: Shicheng Bi
| | - Yiwen Qu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jianjian Shao
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jianrong Zhang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Weihao Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Li Zhang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jingxuan Ni
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Liting Cao
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
130
|
Anti-Inflammatory Activities of an Anti-Histamine Drug, Loratadine, by Suppressing TAK1 in AP-1 Pathway. Int J Mol Sci 2022; 23:ijms23073986. [PMID: 35409346 PMCID: PMC8999734 DOI: 10.3390/ijms23073986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
Loratadine is an anti-histamine routinely used for treating allergies. However, recent findings have shown that Loratadine may also have anti-inflammatory functions, while their exact mechanisms have not yet been fully uncovered. In this paper, we investigated whether Loratadine can be utilized as an anti-inflammatory drug through a series of in vitro and in vivo experiments using a murine macrophage cell line and an acute gastritis mouse model. Loratadine was found to dramatically reduce the expression of pro-inflammatory genes, including MMP1, MMP3, and MMP9, and inhibit AP-1 transcriptional activation, as demonstrated by the luciferase assay. Therefore, we decided to further explore its role in the AP-1 signaling pathway. The expression of c-Jun and c-Fos, AP-1 subunits, was repressed by Loratadine and, correspondingly, the expression of p-JNK, p-MKK7, and p-TAK1 was also inhibited. In addition, Loratadine was able to reduce gastric bleeding in acute gastritis-induced mice; Western blotting using the stomach samples showed reduced p-c-Fos protein levels. Loratadine was shown to effectively suppress inflammation by specifically targeting TAK1 and suppressing consequent AP-1 signaling pathway activation and inflammatory cytokine production.
Collapse
|
131
|
Yi YS. Potential benefits of ginseng against COVID-19 by targeting inflammasomes. J Ginseng Res 2022; 46:722-730. [PMID: 35399195 PMCID: PMC8979607 DOI: 10.1016/j.jgr.2022.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogenic virus that causes coronavirus disease 2019 (COVID-19), with major symptoms including hyper-inflammation and cytokine storm, which consequently impairs the respiratory system and multiple organs, or even cause death. SARS-CoV-2 activates inflammasomes and inflammasome-mediated inflammatory signaling pathways, which are key determinants of hyperinflammation and cytokine storm in COVID-19 patients. Additionally, SARS-CoV-2 inhibits inflammasome activation to evade the host's antiviral immunity. Therefore, regulating inflammasome initiation has received increasing attention as a preventive measure in COVID-19 patients. Ginseng and its major active constituents, ginsenosides and saponins, improve the immune system and exert anti-inflammatory effects by targeting inflammasome stimulation. Therefore, this review discussed the potential preventive and therapeutic roles of ginseng in COVID-19 based on its regulatory role in inflammasome initiation and the host's antiviral immunity.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, 16227, Republic of Korea.
| |
Collapse
|
132
|
Al-Hussaniy HA, Al-Kuraishy HM, Abdulameer AGA. The Use of Panax Ginseng to Reduce the Cardiotoxicity of Doxorubicin and Study its Effect on Modulating Oxidative Stress, Inflammatory, and Apoptosis Pathways. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND: Doxorubicin (DOX) is a broad-spectrum anti-cancer drug that is used to treat a variety of cancers, including blood cancers such as leukemia and solid tissue cancers. However, its use some time limited because of its cardiotoxicity.
OBJECTIVE: The objective of the study was to determine the cardioprotective effect of ginseng in the case of cardiotoxicity caused by doxorubicin therapy.
Methods: Thirty experimental animals (male Sprague Wistar rats) were used in this research and they were separated into three groups: Rats in Group I (n# = 10) were given distilled water plus normal saline, rats in Group II (n# = 10) were given distilled water plus doxorubicin, and rats in Group III (n# = 10) were given Panax ginseng plus doxorubicin. Serum concentration, malondialdehyde (MDA), glutathione reductase (GSH), lipid peroxidase (LPO), TNF (ng/L), cardiac troponin (ng/L), brain natriuretic peptide BNP(g/L), and caspase-3 (pmol/L) levels were measured in all groups.
RESULTS: Doxorubicin caused substantial cardiotoxicity as a result of a significant increase in the elevation of cTnI to 40.09 ± 6.67 (ng/L). In addition, MDA, LPO, TNF-α, and caspase-3 levels were increased in doxorubicin group compared to the control group p < 0.05. Panax ginseng reduced cardiac troponin (cTnI) However, its effect on reduction of BNP levels insignificantly compared to the doxorubicin group p = 0.06. Panax ginseng reduced LPO and MDA and raised the antioxidant potential biomarker GSH significantly compared to the doxorubicin group p < 0.05. Panax ginseng significantly reduced inflammatory (TNF-α) and apoptotic (caspase-3) biomarkers when compared to the doxorubicin group.
CONCLUSIONS: According to the findings of this study, Panax ginseng suppresses reactive oxygen species and inflammatory and apoptotic pathways in experimental rats, thereby preventing doxorubicin-induced cardiovascular events.
Collapse
|
133
|
Wang H, Wu Z, Liu Y, Wang M, Stalin A, Guo S, Li J, Wu C, Zhang J, Tan Y, Huang Z, Lu S, Fan X, Wu J. A novel strategy to reveal clinical advantages and molecular mechanism of aidi injection in the treatment of pancreatic cancer based on network meta-analysis and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114852. [PMID: 34838619 DOI: 10.1016/j.jep.2021.114852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pancreatic cancer is a common malignancy worldwide due to its poor prognosis and high mortality rate. It is clinically proven that the combination of chemotherapeutic drugs and Traditional Chinese Medicine injections (TCMIs) significantly improves the therapeutic effect. AIM OF THE STUDY To evaluate the efficacy and clinical benefits of TCMIs in combination with chemotherapy in the treatment of pancreatic cancer and to explore the mechanism of clinical advantage of Aidi injection. METHODS Randomized controlled trials (RCTs) were searched in databases by NMA before December 29, 2020. WinBUGS 1.4, Stata 14.0, and R 4.0.4 software were used for calculations. All results were expressed as odds ratios and 95% credible intervals. Through the network pharmacology method, the chemical components and their targets, as well as the disease targets were further analyzed. And then, biological experiments were integrated to verify the results of network pharmacology analysis. (PROSPERO ID: CRD42021283559). RESULTS A total of 33 RCTs with 8 TCMIs and 2011 patients were included. The results of NMA showed that Aidi injection can significantly improve the clinical efficacy (OR = 0.34, 95%CI: 0.16-0.74), and the clinical advantage was that it can significantly alleviate the leukopenia and thrombocytopenia caused by chemotherapy (OR = 5.65, 95%CI: 1.18-28.13). A total of 23 chemical compounds and 280 potential targets for Aidi injection were obtained from the online databases. Among them, there were 22 compounds, 50 targets and 211 signaling pathways closely related to leukopenia. Five genes were predicted to be core targets of ADI in alleviating leukopenia, and 2 of them (TP53 and VEGFA) were confirmed by biological experiments as regulatory targets of ADI in the treatment of PC. CONCLUSIONS In conclusion, TCMIs in combination with chemotherapy, can improve clinical efficacy and safety in the treatment of pancreatic cancer. However, the overall evidence base is low, and large samples with multi-center RCTs are still needed to support further research findings. Aidi injection can alleviate leukopenia mainly by intervening in oxidative stress, regulating cell proliferation and apoptosis, and regulating the inflammatory response. The combined application of NMA, network pharmacology, and biological experiments provides a reference for clinical evaluation and mechanism of action exploration of other drugs.
Collapse
Affiliation(s)
- Haojia Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Miaomiao Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Antony Stalin
- State Key Laboratory of Subtropical Silviculture, Department of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, 311300, China.
| | - Siyu Guo
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jialin Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
134
|
Sottero B, Testa G, Gamba P, Staurenghi E, Giannelli S, Leonarduzzi G. Macrophage polarization by potential nutraceutical compounds: A strategic approach to counteract inflammation in atherosclerosis. Free Radic Biol Med 2022; 181:251-269. [PMID: 35158030 DOI: 10.1016/j.freeradbiomed.2022.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
Chronic inflammation represents a main event in the onset and progression of atherosclerosis and is closely associated with oxidative stress in a sort of vicious circle that amplifies and sustains all stages of the disease. Key players of atherosclerosis are monocytes/macrophages. According to their pro- or anti-inflammatory phenotype and biological functions, lesional macrophages can release various mediators and enzymes, which in turn contribute to plaque progression and destabilization or, alternatively, lead to its resolution. Among the factors connected to atherosclerotic disease, lipid species carried by low density lipoproteins and pro-oxidant stimuli strongly promote inflammatory events in the vasculature, also by modulating the macrophage phenotyping. Therapies specifically aimed to balance macrophage inflammatory state are increasingly considered as powerful tools to counteract plaque formation and destabilization. In this connection, several molecules of natural origin have been recognized to be active mediators of diverse metabolic and signaling pathways regulating lipid homeostasis, redox state, and inflammation; they are, thus, considered as promising candidates to modulate macrophage responsiveness to pro-atherogenic stimuli. The current knowledge of the capability of nutraceuticals to target macrophage polarization and to counteract atherosclerotic lesion progression, based mainly on in vitro investigation, is summarized in the present review.
Collapse
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy.
| |
Collapse
|
135
|
Anti-Inflammatory Functions of Methanol Extract from Malus baccata (L.) Borkh. Leaves and Shoots by Targeting the NF-κB Pathway. PLANTS 2022; 11:plants11050646. [PMID: 35270116 PMCID: PMC8912290 DOI: 10.3390/plants11050646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023]
Abstract
Malus baccata (L.) Borkh. is a widely used medical plant in Asia. Since the anti-inflammatory mechanism of this plant is not fully understood, the aim of this study was to explore the anti-inflammatory function and mechanism of Malus baccata (L.) Borkh. methanol extract (Mb-ME). For in vitro experiments, nitric oxide production assay, PCR, overexpression strategy, immunoblotting, luciferase reporter assay, and immunoprecipitation were employed to explore the molecular mechanism and the target proteins of Mb-ME. For in vivo experiments, an HCl/EtOH-induced gastritis mouse model was used to confirm the anti-inflammatory function. Mb-ME showed a strong ability to inhibit the production of nitric oxide and the expression of inflammatory genes. Mb-ME decreased NF-κB luciferase activity mediated by MyD88 and TRIF. Moreover, Mb-ME blocked the activation of Src, Syk, p85, Akt, p50, p60, IKKα/β, and IκBα in LPS-induced RAW264.7 cells. Overexpression and immunoprecipitation analyses suggested Syk and Src as the target enzymes of Mb-ME. In vitro results showed that Mb-ME could alleviate gastritis and relieve the protein expression of p-Src, p-Syk, and COX-2, as well as the gene expression of COX-2 and TNF-α. In summary, this study implied that Mb-ME performs an anti-inflammatory role by suppressing Syk and Src in the NF-κB signaling pathway, both in vivo and in vitro.
Collapse
|
136
|
Zhang H, Hua X, Zheng D, Wu H, Li C, Rao P, Wen M, Choi YE, Xue Z, Wang Y, Li Y. De Novo Biosynthesis of Oleanane-Type Ginsenosides in Saccharomyces cerevisiae Using Two Types of Glycosyltransferases from Panax ginseng. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:2231-2240. [PMID: 35148079 DOI: 10.1021/acs.jafc.1c07526] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Oleanane-type ginsenosides are highly biologically active substances in Panax ginseng, a popular Chinese dietary plant. Lack of key enzymes for glycosylation reactions has hindered de novo synthesis of these bioactive molecules. We mined candidate glycosyltransferases (GTs) of the ginseng database by combining key metabolites and transcriptome coexpression analyses and verified their function using in vitro enzymatic assays. The PgCSyGT1, a cellulose synthase-like GT rather than a UDP-dependent glucuronosyltransferase (UGT), was verified as the key enzyme for transferring a glucuronosyl moiety to the free C3-OH of oleanolic acid to synthesize calenduloside E. Two UGTs (PgUGT18 and PgUGT8) were first identified as, respectively, catalyzing the glycosylation reaction of the second sugar moiety of C3 and the C28 in the oleanane-type ginsenoside biosynthetic pathway. Then, we integrated these GTs in combinations into Saccharomyces cerevisiae genome and realized de novo biosynthesis of oleanane-type ginsenosides with a yield of 1.41 μg/L ginsenoside Ro in shake flasks. This report provides a basis for effective biosynthesis of diverse oleanane-type ginsenosides in microbial cell factories.
Collapse
Affiliation(s)
- He Zhang
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Harbin 150040, China
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Xin Hua
- College of Life Science, Northeast Forestry University, Harbin 150040, China
- Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, Northeast Forestry University, Harbin 150040, China
| | - Dongran Zheng
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Harbin 150040, China
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Hao Wu
- College of Life Science, Northeast Forestry University, Harbin 150040, China
- Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, Northeast Forestry University, Harbin 150040, China
| | - Chuanwang Li
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Harbin 150040, China
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Pan Rao
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Harbin 150040, China
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Mengliang Wen
- Yunnan Enov Bioengineering Co., Ltd, 2nd Floor, Building B2, 16 PuFa Road, Export Processing Zone, Economic Development Zone, Kunming, Yunnan 650217, China
| | - Yong-Eui Choi
- Department of Forest Resources, College of Forest and Environmental Sciences, Kangwon National University, Chunchon 200-701, Republic of Korea
| | - Zheyong Xue
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Harbin 150040, China
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Yu Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China
- Heilongjiang Key Laboratory of Plant Bioactive Substance Biosynthesis and Utilization, Northeast Forestry University, Harbin 150040, China
| | - Yuhua Li
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration (Northeast Forestry University), Ministry of Education, Harbin 150040, China
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
137
|
Min D, Kim B, Ko SG, Kim W. Effect and Mechanism of Herbal Medicines on Cisplatin-Induced Anorexia. Pharmaceuticals (Basel) 2022; 15:ph15020208. [PMID: 35215322 PMCID: PMC8877473 DOI: 10.3390/ph15020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/04/2022] Open
Abstract
Cisplatin is a well-known chemotherapeutic agent used to treat various types of cancers; however, it can also induce anorexia, which results in reduced food intake, loss of body weight, and lower quality of life. Although drugs such as megestrol acetate and cyproheptadine are used to decrease this severe feeding disorder, they can also induce side effects, such as diarrhea and somnolence, which limit their widespread use. Various types of herbal medicines have long been used to prevent and treat numerous gastrointestinal tract diseases; however, to date, no study has been conducted to analyze and summarize their effects on cisplatin-induced anorexia. In this paper, we analyze 12 animal studies that used either a single herbal medicine extract or mixtures thereof to decrease cisplatin-induced anorexia. Among the herbal medicines, Ginseng Radix was the most used, as it was included in seven studies, whereas both Glycyrrhizae Radix et Rhizoma and Angelicae Gigantis Radix were used in four studies. As for the mechanisms of action, the roles of serotonin and its receptors, cytokines, white blood cells, ghrelin, and leptin were investigated. Based on these results, we suggest that herbal medicines could be considered a useful treatment method for cisplatin-induced anorexia.
Collapse
Affiliation(s)
- Daeun Min
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea;
| | - Bonglee Kim
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 022447, Korea; (B.K.); (S.-G.K.)
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 022447, Korea; (B.K.); (S.-G.K.)
| | - Woojin Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea;
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 022447, Korea; (B.K.); (S.-G.K.)
- Correspondence:
| |
Collapse
|
138
|
de Oliveira Zanuso B, de Oliveira Dos Santos AR, Miola VFB, Gissoni Campos LM, Spilla CSG, Barbalho SM. Panax ginseng and aging related disorders: A systematic review. Exp Gerontol 2022; 161:111731. [PMID: 35143871 DOI: 10.1016/j.exger.2022.111731] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022]
Abstract
The aging process predisposes numerous homeostatic disorders, metabolic disorders, cardiovascular diseases, neurodegenerative diseases, and cancer. Changes in diet and lifestyle and therapeutic adjuvants are essential to minimize the effects of comorbidities associated with aging. Natural products such as Panax ginseng have been used to treat and prevent diseases related to aging. This review aims to investigate the effects of Panax ginseng in various conditions associated with aging, such as inflammation, oxidative stress, mitochondrial dysfunction, apoptosis, neurodegenerative and metabolic disorders, cardiovascular diseases, and cancer. The ginsenosides, chemical constituents found in Panax ginseng, can inhibit the effects of inflammatory cytokines, inhibit signaling pathways that induce inflammation, and inhibit cells that participate in inflammatory processes. Besides, ginsenosides are involved in neuroprotective effects on the central nervous system due to anti-apoptotic, antioxidant, and anti-inflammatory effects. The use of ginseng extract showed actions on lipid homeostasis, positively regulating high-density lipoprotein, down-regulating low-density lipoprotein and triglyceride levels, and producing beneficial effects on vascular endothelial function. The use of this plant in cancer resulted in improved quality of life and mood. It decreased symptoms of fatigue, nausea, vomiting, and dyspnea, reducing anxiety. Panax ginseng has been shown to exert potent therapeutic benefits that can act as a complementary treatment in managing patients with chronic diseases related to aging.
Collapse
Affiliation(s)
- Bárbara de Oliveira Zanuso
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil
| | - Ana Rita de Oliveira Dos Santos
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil
| | - Vitor Fernando Bordin Miola
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil
| | - Leila M Gissoni Campos
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil
| | - Caio Sergio Galina Spilla
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Higino Muzzi Filho, 1001, Marília, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marilia (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, São Paulo, Brazil; Department of Biochemistry and Nutrition, Faculty of Food Technology of Marília, Marília, São Paulo, Brazil.
| |
Collapse
|
139
|
Pharmacokinetics of Ginsenoside Rb1, Rg3, Rk1, Rg5, F2, and Compound K from Red Ginseng Extract in Healthy Korean Volunteers. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8427519. [PMID: 35111231 PMCID: PMC8803428 DOI: 10.1155/2022/8427519] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 12/24/2021] [Accepted: 01/04/2022] [Indexed: 11/17/2022]
Abstract
Individual differences in ginsenoside pharmacokinetics following ginseng administration in humans are still unclear. We aimed to investigate the pharmacokinetic properties of various ginsenosides, including Rb1, Rg3, Rg5, Rk1, F2, and compound K (CK), after a single oral administration of red ginseng (RG) and bioconverted red ginseng extract (BRG). This was a randomized, open-label, single-dose, single-sequence crossover study with washout every 1 week, and 14 healthy Korean men were enrolled. All subjects were equally assigned to two groups and given RG or BRG capsules. The pharmacokinetic parameters of ginsenosides were measured from the plasma drug concentration-time curve of individual subjects. Ginsenosides Rg3, Rk1 + Rg5, F2, and CK in the BRG group showed a higher C max, AUC(0-t), and AUC(0-∞) and shorter T max (for CK) than those in the RG group. These results suggest that BRG may lead to a higher absorption rate of bioactive ginsenosides. This study provides valuable information on the pharmacokinetics of various bioactive ginsenosides, which is needed to enhance the therapeutic efficacy and pharmacological activity of ginseng.
Collapse
|
140
|
Korean red ginseng saponin fraction exerts anti-inflammatory effects by targeting the NF-κB and AP-1 pathways. J Ginseng Res 2022; 46:489-495. [PMID: 35600780 PMCID: PMC9120761 DOI: 10.1016/j.jgr.2022.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
Abstract
Background Although ginsenosides and saponins in Korea red ginseng (KRG) shows various pharmacological roles, their roles in the inflammatory response are little known. This study investigated the anti-inflammatory role of ginsenosides identified from KRG saponin fraction (RGSF) and the potential mechanism in macrophages. Methods The ginsenoside composition of RGSF was identified by high-performance liquid chromatography (HPLC) analysis. An anti-inflammatory effect of RGSF and its mechanisms were studied using nitric oxide (NO) and prostaglandin E2 (PGE2) production assays, mRNA expression analyses of inflammatory genes and cytokines, luciferase reporter gene assays of transcription factors, and Western blot analyses of inflammatory signaling pathways using the lipopolysaccharide (LPS)-treated RAW264.7 cells. Results HPLC analysis identified the types and amounts of various panaxadiol ginsenosides in RGSF. RGSF reduced the generation of inflammatory molecules and mRNA levels of inflammatory enzymes and cytokines in LPS-treated RAW264.7 cells. Additionally, RGSF inhibited the signaling pathways of NF-κB and AP-1 by suppressing both transcriptional factors and signaling molecules in LPS-treated RAW264.7 cells. Conclusion RGSF contains ginsenosides that have anti-inflammatory action via restraining the NF-κB and AP-1 signaling pathways in macrophages during inflammatory responses.
Collapse
|
141
|
Lu Q, Li R, Yang Y, Zhang Y, Zhao Q, Li J. Ingredients with anti-inflammatory effect from medicine food homology plants. Food Chem 2022; 368:130610. [PMID: 34419798 DOI: 10.1016/j.foodchem.2021.130610] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/18/2021] [Accepted: 07/13/2021] [Indexed: 02/09/2023]
Abstract
Inflammation occurs when the immune system responses to external harmful stimuli and infection. Chronic inflammation induces various diseases. A variety of foods are prescribed in the traditional medicines of many countries all over the world, which gave birth to the concept of medicine food homology. Over the past few decades, a number of secondary metabolites from medicine food homology plants have been demonstrated to have anti-inflammatory effects. In the present review, the effects and mechanisms of the medicine food homology plants-derived active components on relieving inflammation and inflammation-mediated diseases were summarized and discussed. The information provided in this review is valuable to future studies on anti-inflammatory ingredients derived from medicine food homology plants as drugs or food supplements.
Collapse
Affiliation(s)
- Qiuxia Lu
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China
| | - Rui Li
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China
| | - Yixi Yang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China
| | - Yujin Zhang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Qi Zhao
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Jian Li
- School of Medicine, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
142
|
Zarneshan SN, Fakhri S, Khan H. Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: A mechanistic approach. Pharmacol Res 2022; 177:106099. [DOI: 10.1016/j.phrs.2022.106099] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/14/2022] [Accepted: 01/23/2022] [Indexed: 12/15/2022]
|
143
|
Sng KS, Li G, Zhou LY, Song YJ, Chen XQ, Wang YJ, Yao M, Cui XJ. Ginseng extract and ginsenosides improve neurological function and promote antioxidant effects in rats with spinal cord injury: A meta-analysis and systematic review. J Ginseng Res 2022; 46:11-22. [PMID: 35058723 PMCID: PMC8753526 DOI: 10.1016/j.jgr.2021.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/26/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) is defined as damage to the spinal cord that temporarily or permanently changes its function. There is no definite treatment established for neurological complete injury patients. This study investigated the effect of ginseng extract and ginsenosides on neurological recovery and antioxidant efficacies in rat models following SCI and explore the appropriate dosage. Searches were done on PubMed, Embase, and Chinese databases, and animal studies matches the inclusion criteria were selected. Pair-wise meta-analysis and subgroup analysis were performed. Ten studies were included, and the overall methodological qualities were low quality. The result showed ginseng extract and ginsenosides significantly improve neurological function, through the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale (pooled MD = 4.40; 95% CI = 3.92 to 4.88; p < 0.00001), significantly decrease malondialdehyde (MDA) (n = 290; pooled MD = −2.19; 95% CI = −3.16 to −1.22; p < 0.0001) and increase superoxide dismutase (SOD) levels (n = 290; pooled MD = 2.14; 95% CI = 1.45 to 2.83; p < 0.00001). Both low (<25 mg/kg) and high dosage (≥25 mg/kg) showed significant improvement in the motor function recovery in SCI rats. Collectively, this review suggests ginseng extract and ginsenosides has a protective effect on SCI, with good safety and a clear mechanism of action and may be suitable for future clinical trials and applications.
Collapse
|
144
|
Ratan ZA, Rabbi Mashrur F, Runa NJ, Kwon KW, Hosseinzadeh H, Cho JY. Ginseng, a promising choice for SARS-COV-2: A mini review. J Ginseng Res 2022; 46:183-187. [PMID: 35095288 PMCID: PMC8783644 DOI: 10.1016/j.jgr.2022.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 12/26/2022] Open
|
145
|
Shan M, Zhang X, Fang X, Tian J, Song L, Chen Y, Qiu Z, Zhu D, Luo H, Wang Z. Structural analysis of Panax ginseng glycoproteins and its anti-oligoasthenozoospermia effect in vivo. Int J Biol Macromol 2021; 193:778-788. [PMID: 34743938 DOI: 10.1016/j.ijbiomac.2021.10.136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022]
Abstract
A component from ginseng in which sugars and proteins are covalently bound is named Panax ginseng glycoproteins (PGG). The contents of neutral carbohydrate, acid carbohydrate, and protein were 45.4%, 4.3% and 51.1%. The average molecular weight was 12,690 Da. The structure analysis showed that PGG had more than 1100 glycoproteins with molecular weight between 308.13 Da and 9991.52 Da, it was divided into two parts: long chain structure and short chain structure. These two parts were compared in molecular mass, number of amino acids, theoretical pI, instability index, aliphatic index and GRAVY. The in vivo distribution test of mice showed that PGG was enriched in mice testis, testicular tissue sections showed strong fluorescence signal expression on the surface of seminiferous tubules. We used cyclophosphamide (CP) to establish a mice model of oligoasthenozoospermia to investigate the anti-oligoasthenozoospermic effect of PGG. The results showed that PGG increased the levels of sex hormones T, FSH, PRL and sperm quality. Histopathology demonstrated that PGG promoted the differentiation process. The organ coefficient indicated that PGG had no obvious toxic and side effects. And the mechanism may be to affect the expression of protein levels such as p-ERK/ERK, p-AKT/AKT, Caspase-3, Bcl-2 and Bax. Therefore, PGG has the potential to develop into drugs for improving spermatogenic disorders.
Collapse
Affiliation(s)
- Mengyao Shan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; The first hospital of Jilin university, Changchun 130117, China
| | - Xiaoying Zhang
- The first hospital of Jilin university, Changchun 130117, China
| | - Xiaoxue Fang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jianming Tian
- Jilin Academy of Chinese Medicine and Material Medica Science, Changchun 130012, China
| | - Lianlian Song
- Jilin Academy of Chinese Medicine and Material Medica Science, Changchun 130012, China
| | - Yinghong Chen
- Jilin Academy of Chinese Medicine and Material Medica Science, Changchun 130012, China
| | - Zhidong Qiu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Difu Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Haoming Luo
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Zhe Wang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
146
|
Ginsenoside Rg1 Prevents Cognitive Impairment and Hippocampal Neuronal Apoptosis in Experimental Vascular Dementia Mice by Promoting GPR30 Expression. Neural Plast 2021; 2021:2412220. [PMID: 34899899 PMCID: PMC8664545 DOI: 10.1155/2021/2412220] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/19/2021] [Indexed: 01/02/2023] Open
Abstract
This study is aimed at investigating the potential roles of G protein-coupled estrogen receptor 1 (GPER, also known as GPR30) in the preventive effect of ginsenoside Rg1 against cognitive impairment and hippocampal cell apoptosis in experimental vascular dementia (VD) in mice. The effects of bilateral common carotid artery stenosis (BCAS) on GPR30 expression at mRNA level were evaluated. Thereafter, the BCAS mouse model was utilized to evaluate the protection of Rg1 (0.1, 1, 10 mg/kg, 14 days, ip). Spatial memory was evaluated by water Morris Maze 7 days post BCAS. After behavioral tests, neuronal apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and potential mechanisms were determined using western blotting and quantitative real-time PCR. Our results showed that GPR30 expression in the hippocampal region at mRNA level was promoted 30 min, 3 h, 6 h, and 24 h following BCAS. Ginsenoside Rg1 (1 or 10 mg/kg, 14 days, ip) promoted GPR30 expression in the hippocampus of model mice (after behavioral tests) but did not alter GPR30 expression in the hippocampus of control mice. Moreover, treatment of ginsenoside Rg1 (10 mg/kg) or G1 (5 μg/kg), a GPR30 agonist, prevented BCAS-induced memory impairment and hippocampal neuronal loss and apoptosis and promoted the ratio of Bcl-2 to Bax expression in the hippocampus (after behavioral tests). On the contrary, G15 (185 μg/kg), an antagonist of GPR30, aggravated BCAS-induced hippocampal neuronal loss and apoptosis. Finally, drug-target molecular docking pointed that Rg1 had a lower binding energy with GPR30 compared with Bax and Bcl-2. Together, our data implicate that ginsenoside Rg1 prevents cognitive impairment and hippocampal neuronal apoptosis in VD mice, likely through promoting GPR30 expression. These results would provide important implications for the application of Rg1 in the treatment of VD.
Collapse
|
147
|
Qu B, Cao T, Wang M, Wang S, Li W, Li H. Ginsenosides Rd monomer inhibits proinflammatory cytokines production and alleviates DSS-colitis by NF-κB and P38MAPK pathways in mice. Immunopharmacol Immunotoxicol 2021; 44:110-118. [PMID: 34898349 DOI: 10.1080/08923973.2021.2012482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Ulcerative colitis (UC) is dramatically increasing worldwide, cannot be thoroughly cured, and reduces patients' quality of life. Excessive activation of macrophages and over-production of cytokines play an important role in the pathogenesis of UC. Therefore, for its treatment, inhibiting macrophages' hyperactivation would be effective to develop new treatment approaches. Ginsenosides, extracted from ginseng, show an anti-inflammatory effect on the immunologic process. Our study used ginsenosides Rd monomer (GRd) to intervene in DSS-induced colitis mouse models and tested the immunological effect of macrophages. METHOD We observed body weights, weights of colons, colonic lengths, and inflammatory scores, as well as histological changes of DSS/DSS-GRd mice. We also isolated intestinal and peritoneal macrophages, performed qRT-PCR and ELISA to detect cytokines production by macrophages, and screened possible involved pathways by Western blotting. RESULTS Administering 20 mg/Kg GRd to DSS mice for 7-14 days reduced colonic inflammation. Moreover, both in vivo and in vitro, levels of TNF-α, IFN-γ, IL-6, IL-12/23p40, and IL-17A were all inhibited by GRd at 14 days in intestinal macrophages, and 20 μmol/L GRd at 12 h in peritoneal macrophages, respectively, but longer time made no more benefit. Western blotting showed GRd could decrease expression of pJNK, p-p38, pIκBα, and P65 in nuclear. CONCLUSIONS Our data indicate that GRd could down-regulate cytokines production in macrophages and alleviate DSS-colitis in mice, which may be related to NF-κB and P38MAPK pathways. These results suggest that GRd has an anti-inflammatory effect on experimental colitis and may have potential efficacy in the treatment of UC alone or in combination.
Collapse
Affiliation(s)
- Bo Qu
- Digestive Department, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ting Cao
- Digestive Department, The 3rd Affiliated Hospital of Qiqihar Medical College, Qiqihar, China
| | - Miao Wang
- Digestive Department, Yiwu Central Hospital, Yiwu, China
| | - Shuang Wang
- Digestive Department, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wanying Li
- Digestive Department, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Li
- Digestive Department, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
148
|
You L, Cha S, Kim MY, Cho JY. Ginsenosides are active ingredients in Panax ginseng with immunomodulatory properties from cellular to organismal levels. J Ginseng Res 2021; 46:711-721. [DOI: 10.1016/j.jgr.2021.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
|
149
|
Arafa ESA, Refaey MS, Abd El-Ghafar OAM, Hassanein EHM, Sayed AM. The promising therapeutic potentials of ginsenosides mediated through p38 MAPK signaling inhibition. Heliyon 2021; 7:e08354. [PMID: 34825082 PMCID: PMC8605069 DOI: 10.1016/j.heliyon.2021.e08354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/06/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022] Open
Abstract
The p38 mitogen-activated protein kinases (p38 MAPK) is a 38kD polypeptide recognized as the target for many potential anti-inflammatory agents. Accumulating evidence indicates that p38 MAPK could perform many roles in human disease pathophysiology. Therefore, great therapeutic benefits can be attained from p38 MAPK inhibitors. Ginseng is an exceptionally valued medicinal plant of the family Araliaceae (Panax genus). Recently, several studies targeted the therapeutic effects of purified individual ginsenoside, the most significant active ingredient of ginseng, and studied its particular molecular mechanism(s) of action rather than whole-plant extracts. Interestingly, several ginsenosides: ginsenosides compound K, F1, Rb1, Rb3, Rc, Rd, Re, Rf, Rg1, Rg2, Rg3, Rg5, Rh1, Rh2, Ro, notoginsenoside R1, and protopanaxadiol have shown to possess great therapeutic potentials mediated by their ability to downregulate p38 MAPK signaling in different cell lines and experimental animal models. Our review compiles the research findings of various ginsenosides as potent anti-inflammatory agents, highlighting the crucial role of p38 MAPK suppression in their pharmacological actions. In addition, in silico studies were conducted to explore the probable binding of these ginsenosides to p38 MAPK. The results obtained proposed p38 MAPK involvement in the beneficial pharmacological activities of ginsenosides in different ailments. p38 MAPK plays many roles in human disease pathophysiology. Therefore, great therapeutic benefits can be attained from p38 MAPK inhibitors. Several ginsenosides showed to possess great therapeutic potentials mediated by its ability to downregulate p38 MAPK signaling. in silico studies were conducted to explore the binding of these ginsenosides to p38 MAPK and evidenced the promising their inhibitory effect.
Collapse
Affiliation(s)
- El-Shaimaa A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates.,Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed S Refaey
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufiya, 32958, Egypt
| | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
150
|
Kim SJ, Lee J, Choi WS, Kim HJ, Kim MY, Kim SC, Kim HS. Ginsenoside F1 Attenuates Eosinophilic Inflammation in Chronic Rhinosinusitis by Promoting NK Cell Function. J Ginseng Res 2021; 45:695-705. [PMID: 34764724 PMCID: PMC8569323 DOI: 10.1016/j.jgr.2021.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Background Ginsenosides have beneficial effects on several airway inflammatory disorders primarily through glucocorticosteroid-like anti-inflammatory activity. Among inflammatory cells, eosinophils play a major pathogenic role in conferring a risk of severe refractory diseases including chronic rhinosinusitis (CRS). However, the role of ginsenosides in reducing eosinophilic inflammation and CRS pathogenesis is unexplored. Methods We investigated the therapeutic efficacy and underlying mechanism of ginsenoside F1 (G-F1) in comparison with those of dexamethasone, a representative glucocorticosteroid, in a murine model of CRS. The effects of G-F1 or dexamethasone on sinonasal abnormalities and infiltration of eosinophils and mast cells were evaluated by histological analyses. The changes in inflammatory cytokine levels in sinonasal tissues, macrophages, and NK cells were assessed by qPCR, ELISA, and immunohistochemistry. Results We found that G-F1 significantly attenuated eosinophilic inflammation, mast cell infiltration, epithelial hyperplasia, and mucosal thickening in the sinonasal mucosa of CRS mice. Moreover, G-F1 reduced the expression of IL-4 and IL-13, as well as hematopoietic prostaglandin D synthase required for prostaglandin D2 production. This therapeutic efficacy was associated with increased NK cell function, without suppression of macrophage inflammatory responses. In comparison, dexamethasone potently suppressed macrophage activation. NK cell depletion nullified the therapeutic effects of G-F1, but not dexamethasone, in CRS mice, supporting a causal link between G-F1 and NK cell activity. Conclusion Our results suggest that potentiating NK cell activity, for example with G-F1, is a promising strategy for resolving eosinophilic inflammation in CRS.
Collapse
Affiliation(s)
- So Jeong Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinju Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Sun Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Stem Cell Immunomodulation Research Center (SCIRC), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyo Jeong Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Stem Cell Immunomodulation Research Center (SCIRC), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi-Yeon Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hun Sik Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Stem Cell Immunomodulation Research Center (SCIRC), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|