1
|
Liu X, Feng L, Jin X, Sun K, Sun Y, Wu X, Xia M, Tan R, Yang H, Li G, Wu W, Zu S, Li A, Chen C, Wan X. Ginsenoside CK Promotes Cell Apoptosis via Regulating P53 Signal Pathway in Non-Small Cell Lung Cancer. Drug Dev Res 2025; 86:e70050. [PMID: 39991943 DOI: 10.1002/ddr.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 02/25/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a malignant tumor with high morbidity and mortality. Ginsenosides have been shown to have strong antitumor activity, inhibiting tumor cell growth and promoting apoptosis. In this paper, the effects of ginsenoside CK on the proliferation and apoptosis of NSCLC 95D and NCI-H460 cells were investigated by CCK8, colony formation assay, flow cytometry, fluorescence staining assay, and Western Blot, and it was found that ginsenoside CK could significantly inhibit the growth and proliferation of non-small cell lung cancer, and it was also clarified that the mechanism of its action was realized by the mitochondrial apoptosis pathway. It provides new therapeutic ideas for lung cancer and other major tumor diseases.
Collapse
Affiliation(s)
- Xinze Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Lin Feng
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, China
| | - Kaijing Sun
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Sun
- Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xinmin Wu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Mingjie Xia
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Renbo Tan
- Northeast Asia Institute of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongmei Yang
- Northeast Asia Institute of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Guangzhe Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Zu
- Jilin Aodong Pharmaceutial Group Co., Ltd Post-Doctoral Research Center, Yanbian, China
| | - Anning Li
- Jilin Aodong Pharmaceutial Group Co., Ltd Post-Doctoral Research Center, Yanbian, China
| | - Changbao Chen
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Xilin Wan
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
- Jilin Aodong Pharmaceutial Group Co., Ltd Post-Doctoral Research Center, Yanbian, China
| |
Collapse
|
2
|
Lu Q, Zhang Z, Liu S, Wang J, Yang X, Yan T, Yang Y, Chen X, Li L, Liu G, Du J, Cao Z. Inhibition of stemness and PD-L1 expression by Pien Tze Huang enhances T cell-mediated killing of colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119447. [PMID: 39914691 DOI: 10.1016/j.jep.2025.119447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pien Tze Huang (PZH) is a traditional medicinal formula consisted of four traditional Chinese medicines (TCMs) including Panax notoginseng (Burk.) F. H. Chen, Snake Gall, Calculus Bovis and Moschus, with clinical efficacy against Colorectal Cancer (CRC). However, the molecular and functional mechanisms underlying this efficacy are not fully elucidated. AIMS OF THE STUDY This study aimed to assess the impact of PZH on CRC cancer stem cells (CSCs), and evaluate the coordination effect of PZH on T cell-mediated anti-CRC with patient-derived autologous T cell co-culture. MATERIALS AND METHODS High-performance liquid chromatography (HPLC) was used to identify the main components of PZH. CCK8 and spheroid formation assays were conducted for assessing cell viability and stemness function. Western blot, immunofluorescence and immunohistochemistry were used to evaluate CSC markers and PD-L1 expression. T cell successful expansion was validated by flow cytometry. Co-culture assay was conducted to explore the activation effect of PZH on T cells. The potential mechanism of PZH in CRC was identified with transcriptomics sequencing and network pharmacology analysis. RESULTS PZH reduced cell viability and spheroid formation ability in CRC, and suppressed the expression of CSC markers - LGR5, DCLK1, and CD133. Moreover, PZH enhanced T cell-mediated cytotoxicity against CRC cells by decreasing the expression of PD-L1. Furthermore, PZH with anti-PD-1 immunotherapy enhancing antitumor efficacy and increasing CD8+ T cell infiltration with decreasing expression of CSC markers and PD-L1. Notably, PZH inhibited CRC patient-derived organoids (PDOs) tumorigenesis and increased autologous T cell cytotoxicity against PDOs (n = 5). Consistently, PZH decreased expression of CSC markers and PD-L1 in PDOs. RNA sequencing and network pharmacology also highlighted that PZH inhibited CRC stemness and PD-L1 to enhance T cell-mediated antitumor effects. CONCLUSIONS PZH enhances T cell-mediated killing by inhibiting the expression of CRC stem cell markers and PD-L1, which warrant further investigation and clinical applications.
Collapse
Affiliation(s)
- Qin Lu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Zhuqing Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Sihan Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Jun Wang
- Department of General Surgery, Second Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China.
| | - Xiaoting Yang
- Talent Research Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Ting Yan
- Department of General Surgery, Second Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China.
| | - Yuping Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Xuzheng Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Li Li
- Department of Health Management, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, 350001, China.
| | - Guanghui Liu
- Department of Ophthalmology, Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, China.
| | - Jian Du
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Zhiyun Cao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China; Key Laboratory of Integrative Medicine, Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
3
|
Wang D, Jin Y, Wang H, Zhang C, Li Y, Subramaniyam S, Sohng JK, Baek NI, Kim YJ. Biosynthesis of a Novel Ginsenoside with High Anticancer Activity by Recombinant UDP-Glycosyltransferase and Characterization of Its Biological Properties. Molecules 2025; 30:898. [PMID: 40005208 PMCID: PMC11858633 DOI: 10.3390/molecules30040898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
UDP-glycosyltransferases (UGTs) contribute to catalyzing the glycosylation of numerous functional natural products and novel derivatives with improved bioactivities. UDP-glucose sterol glucosyltransferase (SGT) is normally involved in the synthesis of sterol glycosides in a variety of organisms. SGT was derived from Salinispora tropica CNB-440 and heterologously expressed in Escherichia coli BL21 (DE3). Novel 12-O-glucosylginsenoside Rh2 was identified using HPLC, high-resolution MS (HR-MS), and NMR analysis. The cell viability assay was performed on 12-O-glucosylginsenoside-treated AGS stomach cancer, HeLa cervical cancer, U87MG glioma, and B16F10 melanoma cell lines. Protein structure modeling, molecular docking, and dynamics simulations were performed using AutoDock 4.2 and GROMACS 2020.1 software. The SGT gene is comprised of 1284 nucleotides and codes for 427 amino acids. The 12-O-glucosylginsenoside Rh2 may be a potential anticancer agent due to its potent viability inhibition of cancer cells. Structural analysis showed critical perspectives into the intermolecular interactions, stability, and binding energetics of the enzyme-ligand complex, with outcomes complementing the experimental data, thereby deepening our understanding of the structural basis of SGT-mediated glycosylation and its functional implications. This report presents a novel ginsenoside, 12-O-glucosylginsenoside Rh2, utilizing reshuffled SGT derived from S. tropica, and provides a promising candidate for anticancer drug research and development.
Collapse
Affiliation(s)
- Dandan Wang
- College of Life Sciences, Yantai University, Yantai 264005, China; (D.W.); (H.W.); (C.Z.); (Y.L.)
| | - Yan Jin
- School of Life Science, Nantong University, Nantong 226019, China;
| | - Hongtao Wang
- College of Life Sciences, Yantai University, Yantai 264005, China; (D.W.); (H.W.); (C.Z.); (Y.L.)
| | - Chenwei Zhang
- College of Life Sciences, Yantai University, Yantai 264005, China; (D.W.); (H.W.); (C.Z.); (Y.L.)
| | - Yao Li
- College of Life Sciences, Yantai University, Yantai 264005, China; (D.W.); (H.W.); (C.Z.); (Y.L.)
| | | | - Jae-Kyung Sohng
- Department of Pharmaceutical Engineering, Institute of Biomolecule Reconstruction, SunMoon University, Asan-si 31460, Chungnam, Republic of Korea;
| | - Nam-In Baek
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea;
| | - Yeon-Ju Kim
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea;
| |
Collapse
|
4
|
Shen Y, Gao Y, Yang G, Zhao Z, Zhao Y, Gao L, Li S. Anti-colorectal cancer effect of total minor ginsenosides produced by lactobacilli transformation of major ginsenosides by inducing apoptosis and regulating gut microbiota. Front Pharmacol 2025; 15:1496346. [PMID: 39845805 PMCID: PMC11750747 DOI: 10.3389/fphar.2024.1496346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Objective Minor ginsenosides have demonstrated promising anticancer effects in previous reports. Total minor ginsenosides (TMG) were obtained through the fermentation of major ginsenosides with Lactiplantibacillus plantarum, and potential anticancer effects of TMGs on the mouse colon cancer cell line CT26.WT, in vitro and in vivo, were investigated. Materials and Methods We employed the Cell Counting Kit-8 (CCK-8), TdT-mediated dUTP nick end labeling (TUNEL), and Western blot analysis in vitro to explore the anti-proliferative and pro-apoptotic functions of TMG in CT26.WT cells. In vivo, a xenograft model was established by subcutaneously injecting mice with CT26.WT cells and administering a dose of 100 mg/kg/day TMG to the tumor-bearing mice. The level of apoptosis and expression of various proteins in the tumor tissues were detected by immunohistochemistry and Western blot. High-throughput 16S rRNA sequencing was used to determine the alterations in the gut microbiota. Results In vitro studies demonstrated that TMG significantly inhibited proliferation and promoted apoptosis in CT26.WT cells. Interestingly, TMG induced apoptosis in CT26.WT cells by affecting the Bax/Bcl-2/caspase-3 pathway. Furthermore, the result of the transplanted tumor model indicated that TMG substantially enhanced the activities of Bax and caspase-3, reduced the activity of Bcl-2, and suppressed the expression of Raf/MEK/ERK protein levels. Fecal analysis revealed that TMG reconstructed the gut microbiota in colorectal cancer-affected mice by augmenting the abundance of the advantageous bacterium Lactobacillus and decreasing the abundance of the harmful bacterium Proteus. Conclusion TMG can exhibit potent anti-colorectal cancer effects through diverse apoptotic mechanisms, with their mode of action closely related to the regulation of gut microbiota.
Collapse
Affiliation(s)
- Yunjiao Shen
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
- School of Chinese Materia Medica, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yansong Gao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Ge Yang
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Zijian Zhao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Yujuan Zhao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Lei Gao
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| | - Shengyu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, China
| |
Collapse
|
5
|
Lin L, Chen D, Li S, Wang T. Ginsenoside Rg1 inhibits multiple myeloma and overcomes bortezomib resistance through AMPK-mTOR pathway. Heliyon 2024; 10:e33935. [PMID: 39071579 PMCID: PMC11283037 DOI: 10.1016/j.heliyon.2024.e33935] [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: 04/16/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Background The resistance of multiple myeloma (MM) to bortezomib (BTZ) has brought multiple challenges to its clinical use. Numerous ginsenosides have potential anti-tumor effects, however, the research on the role of Rg1 in MM has not been reported. Objective To examine the inhibitory impact of Rg1 on the growth of MM and reduce the drug resistance of MM to BTZ through in vivo and in vitro experiments, and to explore their potential mechanism. Methods BTZ drug-resistant cell line RPMI8226R was constructed. Mouse tumor-bearing model was developed by abdominal subcutaneous injection of MM cells. MM cells were treated with AMPK inhibitor Compound C or autophagy inhibitor Chloroquine together with Rg1. RPMI8226R cells were treated with BTZ and Rg1. Cell multiplication was detected using Methylthiazolyldiphenyl-tetrazolium bromide assay. Apoptosis was assessed using flow cytometry. Immunofluorescence assay was employed to assess the autophagy markers LC3. Western blot was utilized to assess the protein expression. Immunohistochemistry was used to detect cell proliferation and apoptosis in tumor tissues. Results In vitro experiments demonstrated that Rg1 could hinder the proliferation of MM cells, promote apoptosis and enhance autophagy. Rg1 could also increase the sensitivity of RPMI8226R to BTZ. In vivo experiments illustrated that Rg1 could hinder the development of MM cells in mice, weaken the proliferation of tumor cells and enhance their apoptosis. Further study found that the anti-MM impact of Rg1 was linked to AMPK-mTOR pathway, the autophagy degree of RPMI8226R was higher than that of RPMI8226, and that Rg1 could inhibit MM and overcome drug resistance through autophagy induced by AMPK-mTOR pathway. Conclusion Rg1 has significant anti-MM effect and can overcome BTZ resistance, and its potential mechanism is related to the regulation of autophagy induced by AMPK-mTOR pathway. Rg1 is a promising adjuvant drug for the treatment of MM.
Collapse
Affiliation(s)
- Li Lin
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Dong Chen
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Shuangyue Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Tiantian Wang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
6
|
Chaurembo AI, Xing N, Chanda F, Li Y, Zhang HJ, Fu LD, Huang JY, Xu YJ, Deng WH, Cui HD, Tong XY, Shu C, Lin HB, Lin KX. Mitofilin in cardiovascular diseases: Insights into the pathogenesis and potential pharmacological interventions. Pharmacol Res 2024; 203:107164. [PMID: 38569981 DOI: 10.1016/j.phrs.2024.107164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/09/2024] [Accepted: 03/29/2024] [Indexed: 04/05/2024]
Abstract
The impact of mitochondrial dysfunction on the pathogenesis of cardiovascular disease is increasing. However, the precise underlying mechanism remains unclear. Mitochondria produce cellular energy through oxidative phosphorylation while regulating calcium homeostasis, cellular respiration, and the production of biosynthetic chemicals. Nevertheless, problems related to cardiac energy metabolism, defective mitochondrial proteins, mitophagy, and structural changes in mitochondrial membranes can cause cardiovascular diseases via mitochondrial dysfunction. Mitofilin is a critical inner mitochondrial membrane protein that maintains cristae structure and facilitates protein transport while linking the inner mitochondrial membrane, outer mitochondrial membrane, and mitochondrial DNA transcription. Researchers believe that mitofilin may be a therapeutic target for treating cardiovascular diseases, particularly cardiac mitochondrial dysfunctions. In this review, we highlight current findings regarding the role of mitofilin in the pathogenesis of cardiovascular diseases and potential therapeutic compounds targeting mitofilin.
Collapse
Affiliation(s)
- Abdallah Iddy Chaurembo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Na Xing
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China.
| | - Francis Chanda
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine (Zhongshan Hospital of Traditional Chinese Medicine), Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui-Juan Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Li-Dan Fu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jian-Yuan Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yun-Jing Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Wen-Hui Deng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao-Dong Cui
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Guizhou Medical University, Guiyang, Guizhou, China
| | - Xin-Yue Tong
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Chi Shu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Food Science College, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China; Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia, Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Kai-Xuan Lin
- Department of Cardiology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine (Zhongshan Hospital of Traditional Chinese Medicine), Zhongshan, Guangdong, China; Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Zhao W, Liu Z, Zhang Z, Chen Z, Liu J, Sun P, Li Y, Qi D, Zhang Z. Si Jun Zi decoction inhibits the growth of lung cancer by reducing the expression of PD-L1 through TLR4/MyD88/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116948. [PMID: 37482260 DOI: 10.1016/j.jep.2023.116948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Si Jun Zi decoction (SJZT) is a traditional Chinese medicine (TCM) formula with the effect of invigorating the spleen qi and replenishing qi. TCM believes that a strong spleen qi helps to strengthen lung qi. Lung cancer is often caused by a deficiency of lung qi. Based on this theory, TCM often applies SJZT to the treatment of lung cancer and has achieved remarkable results. However, the mechanism of SJZT in the treatment of lung cancer remains unclear and requires further study. AIM OF THE STUDY The main purpose of this study is to explore the mechanism of SJZT against lung cancer. MATERIALS AND METHODS In this study, the chemical constituents in SJZT were analyzed by UPLC-Q-Exactive-MS/MS. MTT and cell scratch test were used to determine the cell viability and inhibition of migration in vitro. The effect of SJZT on the expression of PD-L1 protein in A549 cells was detected by Western Blotting (WB). Apoptosis was detected by crystal violet staining. The mouse model of Lewis lung cancer was established in vivo, and the levels of serum TNF-α and IL-2 were detected by enzyme linked immunosorbent assay (ELISA). The protein levels of TLR4, MyD88, NF-κB and PD-L1 in tumor tissues of mice were detected by WB. Quantitative real-time PCR (qRT-PCR) was used to detect the levels of TLR4, MyD88, NF-κB and PD-L1 mRNA. Finally, hematoxylin and eosin (H&E) staining were used to detect the pathological status of tumor tissues in mice. RESULTS A total of 16 active chemical constituents were identified in SJZT. In vitro experiments showed that SJZT could inhibit the growth of A549, induce apoptosis and reduce the expression of PD-L1. In vivo experiments showed that SJZT regulated TLR4/MyD88/NF-κB signaling pathway, decreased the expression of PD-L1, and inhibited tumor growth. CONCLUSIONS SJZT inhibits the growth of lung cancer by regulating TLR4/MyD88/NF-κB signal pathway and reducing the expression of PD-L1.
Collapse
Affiliation(s)
- Wenjie Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China; College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhaidong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhenyong Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zichao Chen
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Jinhua Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Peng Sun
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yaqun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Dongmei Qi
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
8
|
Liu H, Deng R, Zhu CW, Han HK, Zong GF, Ren L, Cheng P, Wei ZH, Zhao Y, Yu SY, Lu Y. Rosmarinic acid in combination with ginsenoside Rg1 suppresses colon cancer metastasis via co-inhition of COX-2 and PD1/PD-L1 signaling axis. Acta Pharmacol Sin 2024; 45:193-208. [PMID: 37749237 PMCID: PMC10770033 DOI: 10.1038/s41401-023-01158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/27/2023] [Indexed: 09/27/2023]
Abstract
Metastasis of colorectal cancer (CRC) is a leading cause of mortality among CRC patients. Elevated COX-2 and PD-L1 expression in colon cancer tissue has been linked to distant metastasis of tumor cells. Although COX-2 inhibitors and immune checkpoint inhibitors demonstrate improved anti-tumor efficacy, their toxicity and variable therapeutic effects in individual patients raise concerns. To address this challenge, it is vital to identify traditional Chinese medicine components that modulate COX-2 and PD-1/PD-L1: rosmarinic acid (RA) exerts striking inhibitory effect on COX-2, while ginsenoside Rg1 (GR) possesses the potential to suppress the binding of PD-1/PD-L1. In this study we investigated whether the combination of RA and GR could exert anti-metastatic effects against CRC. MC38 tumor xenograft mouse model with lung metastasis was established. The mice were administered RA (100 mg·kg-1·d-1, i.g.) alone or in combination with GR (100 mg·kg-1·d-1, i.p.). We showed that RA (50, 100, 150 μM) or a COX-2 inhibitor Celecoxib (1, 3, 9 μM) concentration-dependently inhibited the migration and invasion of MC38 cells in vitro. We further demonstrated that RA and Celecoxib inhibited the metastasis of MC38 tumors in vitro and in vivo via interfering with the COX-2-MYO10 signaling axis and inhibiting the generation of filopodia. In the MC38 tumor xenograft mice, RA administration significantly decreased the number of metastatic foci in the lungs detected by Micro CT scanning; RA in combination with GR that had inhibitory effect on the binding of PD-1 and PD-L1 further suppressed the lung metastasis of colon cancer. Compared to COX-2 inhibitors and immune checkpoint inhibitors, RA and GR displayed better safety profiles without disrupting the tissue structures of the liver, stomach and colon, offering insights into the lower toxic effects of clinical traditional Chinese medicine against tumors while retaining its efficacy.
Collapse
Affiliation(s)
- Huan Liu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng-Wei Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Kuan Han
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Gang-Fan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lang Ren
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhong-Hong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Zhao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Su-Yun Yu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
9
|
Jiang RY, Fang ZR, Zhang HP, Xu JY, Zhu JY, Chen KY, Wang W, Jiang X, Wang XJ. Ginsenosides: changing the basic hallmarks of cancer cells to achieve the purpose of treating breast cancer. Chin Med 2023; 18:125. [PMID: 37749560 PMCID: PMC10518937 DOI: 10.1186/s13020-023-00822-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/16/2023] [Indexed: 09/27/2023] Open
Abstract
In 2021, breast cancer accounted for a substantial proportion of cancer cases and represented the second leading cause of cancer deaths among women worldwide. Although tumor cells originate from normal cells in the human body, they possess distinct biological characteristics resulting from changes in gene structure and function of cancer cells in contrast with normal cells. These distinguishing features, known as hallmarks of cancer cells, differ from those of normal cells. The hallmarks primarily include high metabolic activity, mitochondrial dysfunction, and resistance to cell death. Current evidence suggests that the fundamental hallmarks of tumor cells affect the tissue structure, function, and metabolism of tumor cells and their internal and external environment. Therefore, these fundamental hallmarks of tumor cells enable tumor cells to proliferate, invade and avoid apoptosis. Modifying these hallmarks of tumor cells represents a new and potentially promising approach to tumor treatment. The key to breast cancer treatment lies in identifying the optimal therapeutic agent with minimal toxicity to normal cells, considering the specific types of tumor cells in patients. Some herbal medicines contain active ingredients which can precisely achieve this purpose. In this review, we introduce Ginsenoside's mechanism and research significance in achieving the therapeutic effect of breast cancer by changing the functional hallmarks of tumor cells, providing a new perspective for the potential application of Ginsenoside as a therapeutic drug for breast cancer.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Zi-Ru Fang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Huan-Ping Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Wenzhou Medical University, No. 270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Jun-Yao Xu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jia-Yu Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Ke-Yu Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Zhejiang Chinese Medical University, NO. 548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
| | - Wei Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Wenzhou Medical University, No. 270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Xiao Jiang
- Department of Basic Medical Sciences, Guangxi University of Chinese Medicine, NO. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China.
| | - Xiao-Jia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
10
|
Kim J, Zhang S, Zhu Y, Wang R, Wang J. Amelioration of colitis progression by ginseng-derived exosome-like nanoparticles through suppression of inflammatory cytokines. J Ginseng Res 2023; 47:627-637. [PMID: 37720571 PMCID: PMC10499592 DOI: 10.1016/j.jgr.2023.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/10/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
Background Damage to the healthy intestinal epithelial layer and regulation of the intestinal immune system, closely interrelated, are considered pivotal parts of the curative treatment for inflammatory bowel disease (IBD). Plant-based diets and phytochemicals can support the immune microenvironment in the intestinal epithelial barrier for a balanced immune system by improving the intestinal microecological balance and may have therapeutic potential in colitis. However, there have been only a few reports on the therapeutic potential of plant-derived exosome-like nanoparticles (PENs) and the underlying mechanism in colitis. This study aimed to assess the therapeutic effect of PENs from Panax ginseng, ginseng-derived exosome-like nanoparticles (GENs), in a mouse model of IBD, with a focus on the intestinal immune microenvironment. Method To evaluate the anti-inflammatory effect of GENs on acute colitis, we treated GENs in Caco2 and lipopolysaccharide (LPS) -induced RAW 264.7 macrophages and analyzed the gene expression of pro-inflammatory cytokines and anti-inflammatory cytokines such as TNF-α, IL-6, and IL-10 by real-time PCR (RT-PCR). Furthermore, we further examined bacterial DNA from feces and determined the alteration of gut microbiota composition in DSS-induced colitis mice after administration of GENs through 16S rRNA gene sequencing analysis. Result GENs with low toxicity showed a long-lasting intestinal retention effect for 48 h, which could lead to effective suppression of pro-inflammatory cytokines such as TNF-α and IL-6 production through inhibition of NF-κB in DSS-induced colitis. As a result, it showed longer colon length and suppressed thickening of the colon wall in the mice treated with GENs. Due to the amelioration of the progression of DSS-induced colitis with GENs treatment, the prolonged survival rate was observed for 17 days compared to 9 days in the PBS-treated group. In the gut microbiota analysis, the ratio of Firmicutes/Bacteroidota was decreased, which means GENs have therapeutic effectiveness against IBD. Ingesting GENs would be expected to slow colitis progression, strengthen the gut microbiota, and maintain gut homeostasis by preventing bacterial dysbiosis. Conclusion GENs have a therapeutic effect on colitis through modulation of the intestinal microbiota and immune microenvironment. GENs not only ameliorate the inflammation in the damaged intestine by downregulating pro-inflammatory cytokines but also help balance the microbiota on the intestinal barrier and thereby improve the digestive system.
Collapse
Affiliation(s)
- Jisu Kim
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| | - Shuya Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
| | - Ying Zhu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ruirui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Jang WY, Hwang JY, Cho JY. Ginsenosides from Panax ginseng as Key Modulators of NF-κB Signaling Are Powerful Anti-Inflammatory and Anticancer Agents. Int J Mol Sci 2023; 24:6119. [PMID: 37047092 PMCID: PMC10093821 DOI: 10.3390/ijms24076119] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Nuclear factor kappa B (NF-κB) signaling pathways progress inflammation and immune cell differentiation in the host immune response; however, the uncontrollable stimulation of NF-κB signaling is responsible for several inflammatory illnesses regardless of whether the conditions are acute or chronic. Innate immune cells, such as macrophages, microglia, and Kupffer cells, secrete pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, via the activation of NF-κB subunits, which may lead to the damage of normal cells, including neurons, cardiomyocytes, hepatocytes, and alveolar cells. This results in the occurrence of neurodegenerative disorders, cardiac infarction, or liver injury, which may eventually lead to systemic inflammation or cancer. Recently, ginsenosides from Panax ginseng, a historical herbal plant used in East Asia, have been used as possible options for curing inflammatory diseases. All of the ginsenosides tested target different steps of the NF-κB signaling pathway, ameliorating the symptoms of severe illnesses. Moreover, ginsenosides inhibit the NF-κB-mediated activation of cancer metastasis and immune resistance, significantly attenuating the expression of MMPs, Snail, Slug, TWIST1, and PD-L1. This review introduces current studies on the therapeutic efficacy of ginsenosides in alleviating NF-κB responses and emphasizes the critical role of ginsenosides in severe inflammatory diseases as well as cancers.
Collapse
Affiliation(s)
| | | | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
12
|
Yu HY, Rhim DB, Kim SK, Ban OH, Oh SK, Seo J, Hong SK. Growth promotion effect of red ginseng dietary fiber to probiotics and transcriptome analysis of Lactiplantibacillus plantarum. J Ginseng Res 2023; 47:159-165. [PMID: 36644380 PMCID: PMC9834016 DOI: 10.1016/j.jgr.2022.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 09/15/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Background Red ginseng marc, the residue of red ginseng left after water extraction, is rich in dietary fiber. Dietary fiber derived from fruits or vegetables can promote the proliferation of probiotics, and it is a key technology in the food industry to increase the productivity of probiotics by adding growth-enhancing substances such as dietary fiber. In this study, the effect of red ginseng dietary fiber (RGDF) on the growth of probiotic bacterial strains was investigated at the phenotypic and genetic levels. Methods We performed transcriptome profiling of Lactiplantibacillus plantarum IDCC3501 in two phases of culture (logarithmic (L)-phase and stationary (S)-phase) in two culture conditions (with or without RGDF) using RNA-seq. Differentially expressed genes (DEGs) were identified and classified according to Gene Ontology terms. Results The growth of L.plantarum IDCC3501 was enhanced in medium supplemented with RGDF up to 2%. As a result of DEG analysis, 29 genes were upregulated and 30 were downregulated in the RGDF-treated group in the L-phase. In the S-phase, 57 genes were upregulated and 126 were downregulated in the RGDF-treated group. Among the upregulated genes, 5 were upregulated only in the L-phase, 10 were upregulated only in the S-phase, and 3 were upregulated in both the L- and S-phases. Conclusions Transcriptome analysis could be a valuable tool for elucidating the molecular mechanisms by which RGDF promotes the proliferation of L.plantarum IDCC3501. This growth-promoting effect of RGDF is important, since RGDF could be used as a prebiotic source without additional chemical or enzymatic processing.
Collapse
Affiliation(s)
- Hye-Young Yu
- Laboratory of Red Ginseng Products, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Dong-Bin Rhim
- Laboratory of Red Ginseng Products, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Sang-Kyu Kim
- Laboratory of Red Ginseng Products, Korea Ginseng Corporation, Daejeon, Republic of Korea
- Corresponding author. Sang-Kyu Kim, Laboratory of Red Ginseng Products, Korea Ginseng Corporation, 30, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon, 34128, Republic of Korea.
| | - O-Hyun Ban
- Ildong Bioscience, Pyeongteak, Republic of Korea
| | - Sang-Ki Oh
- Ildong Bioscience, Pyeongteak, Republic of Korea
| | - Jiho Seo
- Laboratory of Red Ginseng Products, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Soon-Ki Hong
- Laboratory of Red Ginseng Products, Korea Ginseng Corporation, Daejeon, Republic of Korea
| |
Collapse
|
13
|
Jeon HJ, You SH, Nam EH, Truong VL, Bang JH, Bae YJ, Rarison RHG, Kim SK, Jeong WS, Jung YH, Shin M. Red ginseng dietary fiber promotes probiotic properties of Lactiplantibacillus plantarum and alters bacterial metabolism. Front Microbiol 2023; 14:1139386. [PMID: 36950168 PMCID: PMC10025373 DOI: 10.3389/fmicb.2023.1139386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Korean red ginseng has been widely used as an herbal medicine. Red ginseng dietary fiber (RGDF) is a residue of the processed ginseng product but still contains bioactive constituents that can be applied as prebiotics. In this study, we evaluated changes on fermentation profiles and probiotic properties of strains that belong to family Lactobacillaceae with RGDF supplementation. Metabolomic analyses were performed to understand specific mechanisms on the metabolic alteration by RGDF and to discover novel bioactive compounds secreted by the RGDF-supplemented probiotic strain. RGDF supplementation promoted short-chain fatty acid (SCFA) production, carbon source utilization, and gut epithelial adhesion of Lactiplantibacillus plantarum and inhibited attachment of enteropathogens. Intracellular and extracellular metabolome analyses revealed that RGDF induced metabolic alteration, especially associated with central carbon metabolism, and produced RGDF-specific metabolites secreted by L. plantarum, respectively. Specifically, L. plantarum showed decreases in intracellular metabolites of oleic acid, nicotinic acid, uracil, and glyceric acid, while extracellular secretion of several metabolites including oleic acid, 2-hydroxybutanoic acid, hexanol, and butyl acetate increased. RGDF supplementation had distinct effects on L. plantarum metabolism compared with fructooligosaccharide supplementation. These findings present potential applications of RGDF as prebiotics and bioactive compounds produced by RGDF-supplemented L. plantarum as novel postbiotic metabolites for human disease prevention and treatment.
Collapse
Affiliation(s)
- Hyeon Ji Jeon
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Hwan You
- Laboratory of Efficacy Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Eoun Ho Nam
- Department of Microbiology, College of Medicine, Inha University, Incheon, Republic of Korea
- Department of Biomedical Sciences, Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea
| | - Van-Long Truong
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Ji-Hong Bang
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yeon-Ji Bae
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Razanamanana H. G. Rarison
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sang-Kyu Kim
- Laboratory of Efficacy Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Woo-Sik Jeong
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Young Hoon Jung
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
- *Correspondence: Young Hoon Jung,
| | - Minhye Shin
- Department of Microbiology, College of Medicine, Inha University, Incheon, Republic of Korea
- Department of Biomedical Sciences, Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea
- Minhye Shin,
| |
Collapse
|
14
|
Ginsenoside Rk3 Inhibits the Extramedullary Infiltration of Acute Monocytic Leukemia Cell via miR-3677-5p/CXCL12 Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3065464. [PMID: 36569343 PMCID: PMC9788880 DOI: 10.1155/2022/3065464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 12/23/2022]
Abstract
Background Acute monocytic leukemia belongs to type M5 of acute myeloid leukemia (AML) classified by FAB, which appears a high incidence of extramedullary infiltration (EMI) and poor prognosis. In this study, we observed the inhibitory effect of ginsenoside Rk3 on the EMI of monocytic leukemia cells and initially explored its related mechanism of targeting the miR-3677-5p/CXCL12 axis. Methods The MTT assay and colony formation assay were used to detect the inhibitory effect of Rk3 on proliferation. Both cellular migration and invasion were observed by the Transwell assay. The expression levels of miR-3677-5p, CXCL12, and CXCR4 were detected by RT-qPCR and Western blot, as well as overexpression of miR-3677-5p by transfected with lentivirus and detection of a dual luciferase reporter gene. The expression of MMP2 and TIMP2 was detected by immunofluorescence. Results Rk3 effectively inhibits the proliferation, migration, and invasion associated with EMI of leukemia. The leukemia cells of M5 patients with EMI showed low expression of miR-3677-5p but high expression of the mRNA of CXCL12 and CXCR4. Overexpression of miR-3677-5p or intervention of CXCL12 effectively inhibited the proliferation, migration, and invasion of SHI-1 cells. The luciferase assay showed that CXCL12 was the downstream target gene of miR-3677-5p. After overexpression of miR-3677-5p or intervention of CXCL12 in combination with Rk3, the inhibitory effect on the proliferation, migration, and invasion of SHI-1 cells was more obvious. Importantly, Rk3 significantly regulated the expression levels of miR-3677-5p, CXCL12, CXCR4, and EMI-related functional proteins including MMP2 and TIMP2. Overexpression of miR-3677-5p or intervention of CXCL12 also regulated the expression of MMP2 and TIMP2. Conclusions The leukemia cells of M5 patients with EMI appeared to have low expression of miR-3677-5p and high expression of the mRNA of CXCL12 and CXCR4, which may be used as indicators of EMI and poor prognosis. Rk3 is effective in inhibiting the EMI of SHI-1 cells by targeting the miR-3677-5p/CXCL12 axis.
Collapse
|
15
|
Todorova V, Ivanov K, Ivanova S. Comparison between the Biological Active Compounds in Plants with Adaptogenic Properties ( Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus and Panax ginseng). PLANTS (BASEL, SWITZERLAND) 2021; 11:64. [PMID: 35009068 PMCID: PMC8747685 DOI: 10.3390/plants11010064] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND In the 1960s, research into plant adaptogens began. Plants with adaptogenic properties have rich phytochemical compositions and have been used by humanity since ancient times. However, it is not still clear whether the adaptogenic properties are because of specific compounds or because of the whole plant extracts. The aim of this review is to compare the bioactive compounds in the different parts of these plants. METHODS The search strategy was based on studies related to the isolation of bioactive compounds from Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus, and Panax ginseng. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. RESULTS This review includes data from 259 articles. The phytochemicals isolated from Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus, and Panax ginseng were described and classified in several categories. CONCLUSIONS Plant species have always played an important role in drug discovery because their effectiveness is based on the hundreds of years of experience with folk medicine in different nations. In our view, there is great potential in the near future for some of the phytochemicals found in these plants species to become pharmaceutical agents.
Collapse
Affiliation(s)
- Velislava Todorova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (K.I.); (S.I.)
| | | | | |
Collapse
|