1
|
Nisar MF, Wan C, Büsselberg D, Calina D, Sharifi-Rad J. Current mechanistic insights into Withaferin A: a promising potential adjuvant anticancer agent from Withania somnifera. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024. [DOI: 10.1007/s00210-024-03662-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
|
2
|
Zhao X, Yuan W, Yang L, Yan F, Cui D. Ginsenoside Rh2 suppresses ferroptosis in ulcerative colitis by targeting specific protein 1 by upregulating microRNA-125a-5p. Eur J Med Res 2024; 29:450. [PMID: 39223620 PMCID: PMC11370063 DOI: 10.1186/s40001-024-02025-w] [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: 12/06/2023] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Worldwide, ulcerative colitis (UC) is becoming increasingly fast growing. Ginsenoside Rh2 has been reported to alleviate UC. However, the latent biological mechanism of Rh2 in the treatment of UC remains uncertain. In this study, the goal was to determine the therapeutic effect of Rh2 on dextran sulfate sodium (DSS)-induced UC. METHODS A DSS-induced UC mouse model was established and divided into 7 groups for Rh2 gavage and/or miR-125a-5p lentivirus injection (n = 10 per group). Colonic specimens were collected for phenotypic and pathological analysis. miR-125a-5p and specific protein 1 (SP1) expression, inflammation-related factors IL-6 and IL-10, and apoptosis were detected in mice. Human normal colon epithelial cell line NCM460 was treated with H2O2 and ferric chloride hexahydrate to construct an in vitro cell model of colitis and induce ferroptosis. Independent sample t-test was used to compare cell proliferation, cell entry, apoptosis, and oxidative stress between the two groups. One way analysis of variance combined with the least significant difference t test was used for comparison between groups. Multiple time points were compared by repeated measurement analysis of variance. RESULTS DSS-induced UC mice had significantly decreased body weight, increased disease activity index, decreased colon length, and decreased miR-125a-5p expression (all P < 0.05). In the DSS-induced mouse model, the expression of miR-125a-5p rebounded and ferroptosis was inhibited after Rh2 treatment (all P < 0.05). Inhibition of miR-125a-5p or upregulation of SP1 expression counteracted the protective effects of Rh2 on UC mice and ferroptosis cell models (all P < 0.05). CONCLUSIONS Rh2 mitigated DSS-induced colitis in mice and restrained ferroptosis by targeting miR-125a-5p. Downregulating miR-125a-5p or elevating SP1 could counteract the protective impacts of Rh2 on ferroptotic cells. The findings convey that Rh2 has a latent application value in the treatment of UC.
Collapse
Affiliation(s)
- Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - WenQiang Yuan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - LiuChan Yang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - Fang Yan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - DeJun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
3
|
Alghareeb SA, Alsughayyir J, Alfhili MA. Ginsenoside Rh2 Regulates the Calcium/ROS/CK1α/MLKL Pathway to Promote Premature Eryptosis and Hemolysis in Red Blood Cells. Toxicol Pathol 2024; 52:284-294. [PMID: 39148410 DOI: 10.1177/01926233241268846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Ginsenoside Rh2 (GRh2) exhibits significant potential as an anticancer agent; however, progress in developing chemotherapeutic drugs is impeded by their toxicity toward off-target tissues. Specifically, anemia caused by chemotherapy is a debilitating side effect and can be caused by red blood cell (RBC) hemolysis and eryptosis. Cells were exposed to GRh2 in the antitumor range and hemolytic and eryptotic markers were examined under different experimental conditions using photometric and cytofluorimetric methods. GRh2 caused Ca2+-independent, concentration-responsive hemolysis in addition to disrupted ion trafficking with K+ and Cl- leakage. Significant increases in cells positive for annexin-V-fluorescein isothiocyanate, Fluo4, and 2,7-dichlorofluorescein were noted upon GRh2 treatment coupled with a decrease in forward scatter and acetylcholinesterase activity. Importantly, the cytotoxic effects of GRh2 were mitigated by ascorbic acid and by blocking casein kinase 1α (CK1α) and mixed lineage kinase domain-like (MLKL) signaling. In contrast, Ca2+ omission, inhibition of KCl efflux, and isosmotic sucrose aggravated GRh2-induced RBC death. In whole blood, GRh2 selectively targeted reticulocytes and lymphocytes. Altogether, this study identified novel mechanisms underlying GRh2-induced RBC death involving Ca2+ buildup, loss of membrane phospholipid asymmetry and cellular volume, anticholinesterase activity, and oxidative stress. These findings shed light on the hematologic toxicity of GRh2 which is crucial for optimizing its utilization in cancer treatment.
Collapse
Affiliation(s)
- Sumiah A Alghareeb
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad A Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Zhang N, Nao J, Zhang S, Dong X. Novel insights into the activating transcription factor 4 in Alzheimer's disease and associated aging-related diseases: Mechanisms and therapeutic implications. Front Neuroendocrinol 2024; 74:101144. [PMID: 38797197 DOI: 10.1016/j.yfrne.2024.101144] [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: 02/05/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Ageing is inherent to all human beings, most mechanistic explanations of ageing results from the combined effects of various physiological and pathological processes. Additionally, aging pivotally contributes to several chronic diseases. Activating transcription factor 4 (ATF4), a member of the ATF/cAMP response element-binding protein family, has recently emerged as a pivotal player owing to its indispensable role in the pathophysiological processes of Alzheimer's disease and aging-related diseases. Moreover, ATF4 is integral to numerous biological processes. Therefore, this article aims to comprehensively review relevant research on the role of ATF4 in the onset and progression of aging-related diseases, elucidating its potential mechanisms and therapeutic approaches. Our objective is to furnish scientific evidence for the early identification of risk factors in aging-related diseases and pave the way for new research directions for their treatment. By elucidating the signaling pathway network of ATF4 in aging-related diseases, we aspire to gain a profound understanding of the molecular and cellular mechanisms, offering novel strategies for addressing aging and developing related therapeutics.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, the Seventh Clinical College of China Medical University, No. 24 Central Street, Xinfu District, Fushun 113000, Liaoning, China.
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| | - Shun Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110000, Liaoning, China.
| |
Collapse
|
5
|
Cao K, Hu S, Wang D, Qiao C, Wang Z, Wang J, Hou W. Clinical efficacy and safety of Chinese herbal injections in combination with platinum-based chemotherapy for advanced non-small cell lung cancer: a systematic review and meta-analysis of 140 randomized controlled trials. Front Oncol 2024; 14:1307836. [PMID: 38371619 PMCID: PMC10869539 DOI: 10.3389/fonc.2024.1307836] [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: 10/05/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
Background and aim Chinese herbal injection (CHI) is a widely used preparation for advanced non-small cell lung cancer (NSCLC) treatment to alleviate the adverse drug reactions and enhance the clinical efficacy of chemotherapy. However, its efficacy and safety in combination with platinum-based chemotherapy (PBC) remain poorly understood owing to the lack of high-level evidence in the face of a wide variety of CHIs. Therefore, in this study, we aimed to explore the efficacy and safety of CHIs in combination with PBC regimens in the treatment of mid- and advanced NSCLC. Methods Systematic evaluation and meta-analysis were conducted as per the Preferred Reporting Project for Systematic Evaluation and Meta-Analysis Protocols (PRISMA-P). Seven databases were comprehensively searched for relevant randomized controlled trials (RCTs) through August 1, 2022. The quality of each study was evaluated based on the Cochrane Handbook for Systematic Reviews of Interventions. Statistical analysis was performed using Revman 5.3, with dichotomies expressed as risk ratio (RR) and 95% confidence interval (CI). Objective response rate (ORR) and disease control rate (DCR) were selected as the primary outcomes, with quality of life (QoL) and toxic side effects as secondary outcomes. Results A total of 140 RCTs were included in this study. The results of the meta-analysis suggested that, compared with PBC alone, PBC combined with CHIs significantly improved the ORR (RR=1.35, 95% CI: 1.30-1.41, P<0.001), DCR (RR=1.15, 95% CI: 1.13-1.18, P<0.001) and QoL (RR=1.29, 95% CI: 1.24-1.33, P<0.001). Moreover, the combination treatment reduced chemotherapy-induced leukopenia (RR=0.69, 95% CI: 0.64-0.75, P<0.001), anemia (RR=0.70, 95% CI: 0.62-0.79, P<0.001), thrombocytopenia (RR=0.68, 95% CI: 0.62-0.75, P<0.001), nausea and vomiting (RR=0.69, 95% CI: 0.63-0.76, P<0.001), diarrhea (RR=0.59, 95% CI: 0.48-0.73, P<0.001), and constipation (RR=0.68, 95% CI: 0.54-0.86, P=0.001). Conclusion According to the available evidence, CHIs in combination with PBC can improve clinical efficacy and reduce the toxic side effects in the treatment of advanced NSCLC. However, considering the study's limitations, more rigorous and high-quality studies are needed to further confirm the results. Systematic review registration https://inplasy.com/inplasy-2022-1-0104/, identifier INPLASY202210104.
Collapse
Affiliation(s)
- Kangdi Cao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuaihang Hu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dandan Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenxi Qiao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuo Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jinkun Wang
- Department of Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Hou
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Yu T, Xu J, Wang Q, Han X, Tu Y, Wang Y, Luo W, Wang M, Liang G. 20(S)-ginsenoside Rh2 inhibits angiotensin-2 mediated cardiac remodeling and inflammation associated with suppression of the JNK/AP-1 pathway. Biomed Pharmacother 2023; 169:115880. [PMID: 37956481 DOI: 10.1016/j.biopha.2023.115880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Enhanced levels of angiotensin-2 (Ang-II) causes hypertensive heart failure (HHF) through non-hemodynamical and hemodynamical alterations. 20(S)-ginsenoside Rh2 (20(S)-Rh2) is a natural ginseng compound with numerous cardiovascular benefits. This investigation elucidates the influence of 20(S)-Rh2 on Ang-II-induced heart failure and cardiac alterations. METHODS Ang-II was administered in C57BL/6 mice for 4 weeks to induce HHF. In the last 2 weeks of treatment, 20(S)-Rh2 was orally administered in mice to assess the potential 20(S)-Rh2 mechanism. Subsequently, RNA sequencing was carried out. RESULTS It was indicated that 20(S)-Rh2 suppresses myocardial fibrosis, hypertrophy, and inflammation, thereby inhibiting cardiac disruption in Ang-II-challenged mice without affecting blood pressure. According to the RNA sequencing data, this cardio-protective effect was linked with the (JNK)/AP 1 pathway. 20(S)-Rh2 alleviated heart tissue and cardiomyocytes inflammation by inhibiting the Ang-II-mediated JNK/AP-1 pathway. Within cardiomyocytes, JNK or AP-1 absence abolished the anti-inflammatory effects of 20(S)-Rh2. CONCLUSION This study investigation indicated that 20(S)-Rh2 prevents cardiovascular dysfunction induced by Ang-II induced by decreasing JNK-regulated inflammatory responses, providing evidence for its use as an efficient regimen for HHF.
Collapse
Affiliation(s)
- Tianxiang Yu
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiachen Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qinyan Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Han
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu Tu
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mengyang Wang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, Jilin132013, China.
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
7
|
Wang L, Zhang Y, Song Z, Liu Q, Fan D, Song X. Ginsenosides: a potential natural medicine to protect the lungs from lung cancer and inflammatory lung disease. Food Funct 2023; 14:9137-9166. [PMID: 37801293 DOI: 10.1039/d3fo02482b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Lung cancer is the malignancy with the highest morbidity and mortality. Additionally, pulmonary inflammatory diseases, such as pneumonia, acute lung injury, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis (PF), also have high mortality rates and can promote the development and progression of lung cancer. Unfortunately, available treatments for them are limited, so it is critical to search for effective drugs and treatment strategies to protect the lungs. Ginsenosides, the main active components of ginseng, have been shown to have anti-cancer and anti-inflammatory activities. In this paper, we focus on the beneficial effects of ginsenosides on lung diseases and their molecular mechanisms. Firstly, the molecular mechanism of ginsenosides against lung cancer was summarized in detail, mainly from the points of view of proliferation, apoptosis, autophagy, angiogenesis, metastasis, drug resistance and immunity. In in vivo and in vitro lung cancer models, ginsenosides Rg3, Rh2 and CK were reported to have strong anti-lung cancer effects. Then, in the models of pneumonia and acute lung injury, the protective effect of Rb1 was particularly remarkable, followed by Rg3 and Rg1, and its molecular mechanism was mainly associated with targeting NF-κB, Nrf2, MAPK and PI3K/Akt pathways to alleviate inflammation, oxidative stress and apoptosis. Additionally, ginsenosides may also have a potential health-promoting effect in the improvement of COPD, asthma and PF. Furthermore, to overcome the low bioavailability of CK and Rh2, the development of nanoparticles, micelles, liposomes and other nanomedicine delivery systems can significantly improve the efficacy of targeted lung cancer treatment. To conclude, ginsenosides can be used as both anti-lung cancer and lung protective agents or adjuvants and have great potential for future clinical applications.
Collapse
Affiliation(s)
- Lina Wang
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Yanxin Zhang
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Zhimin Song
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Qingchao Liu
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Biotechnology & Biomedicine Research Institute, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaoping Song
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
8
|
Wang Y, Han Q, Zhang S, Xing X, Sun X. New perspective on the immunomodulatory activity of ginsenosides: Focus on effective therapies for post-COVID-19. Biomed Pharmacother 2023; 165:115154. [PMID: 37454595 DOI: 10.1016/j.biopha.2023.115154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
More than 700 million confirmed cases of Coronavirus Disease-2019 (COVID-19) have been reported globally, and 10-60% of patients are expected to exhibit "post-COVID-19 symptoms," which will continue to affect human life and health. In the absence of safer, more specific drugs, current multiple immunotherapies have failed to achieve satisfactory efficacy. Ginseng, a traditional Chinese medicine, is often used as an immunomodulator and has been used in COVID-19 treatment as a tonic to increase blood oxygen saturation. Ginsenosides are the main active components of ginseng. In this review, we summarize the multiple ways in which ginsenosides affect post-COVID-19 symptoms, including inhibition of lipopolysaccharide, tumor necrosis factor signaling, modulation of chemokine receptors and inflammasome activation, induction of macrophage polarization, effects on Toll-like receptors, nuclear factor kappa-B, the mitogen-activated protein kinase pathway, lymphocytes, intestinal flora, and epigenetic regulation. Ginsenosides affect virus-mediated tissue damage, local or systemic inflammation, immune modulation, and other links, thus alleviating respiratory and pulmonary symptoms, reducing the cardiac burden, protecting the nervous system, and providing new ideas for the rehabilitation of patients with post-COVID-19 symptoms. Furthermore, we analyzed its role in strengthening body resistance to eliminate pathogenic factors from the perspective of ginseng-epidemic disease and highlighted the challenges in clinical applications. However, the benefit of ginsenosides in modulating organismal imbalance post-COVID-19 needs to be further evaluated to better validate the pharmacological mechanisms associated with their traditional efficacy and to determine their role in individualized therapy.
Collapse
Affiliation(s)
- Yixin Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Qin Han
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Shuxia Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China
| | - Xiaoyan Xing
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College, and Chinese Academy of Medical Sciences, China; Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders,State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, China.
| |
Collapse
|
9
|
Li X, Cao D, Sun S, Wang Y. Anticancer therapeutic effect of ginsenosides through mediating reactive oxygen species. Front Pharmacol 2023; 14:1215020. [PMID: 37564184 PMCID: PMC10411515 DOI: 10.3389/fphar.2023.1215020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Dysregulation of reactive oxygen species (ROS) production and ROS-regulated pathways in cancer cells leads to abnormal accumulation of reactive oxygen species, displaying a double-edged role in cancer progression, either supporting transformation/proliferation and stimulating tumorigenesis or inducing cell death. Cancer cells can accommodate reactive oxygen species by regulating them at levels that allow the activation of pro-cancer signaling pathways without inducing cell death via modulation of the antioxidant defense system. Therefore, targeting reactive oxygen species is a promising approach for cancer treatment. Ginsenosides, their derivatives, and related drug carriers are well-positioned to modulate multiple signaling pathways by regulating oxidative stress-mediated cellular and molecular targets to induce apoptosis; regulate cell cycle arrest and autophagy, invasion, and metastasis; and enhance the sensitivity of drug-resistant cells to chemotherapeutic agents of different cancers depending on the type, level, and source of reactive oxygen species, and the type and stage of the cancer. Our review focuses on the pro- and anticancer effects of reactive oxygen species, and summarizes the mechanisms and recent advances in different ginsenosides that bring about anticancer effects by targeting reactive oxygen species, providing new ideas for designing further anticancer studies or conducting more preclinical and clinical studies.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Siming Sun
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Zhang H, Ma L, Kim E, Yi J, Huang H, Kim H, Raza MA, Park S, Jang S, Kim K, Kim SH, Lee Y, Kim E, Ryoo ZY, Kim MO. Rhein Induces Oral Cancer Cell Apoptosis and ROS via Suppresse AKT/mTOR Signaling Pathway In Vitro and In Vivo. Int J Mol Sci 2023; 24:ijms24108507. [PMID: 37239855 DOI: 10.3390/ijms24108507] [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: 04/03/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Oral cancer remains the leading cause of death worldwide. Rhein is a natural compound extracted from the traditional Chinese herbal medicine rhubarb, which has demonstrated therapeutic effects in various cancers. However, the specific effects of rhein on oral cancer are still unclear. This study aimed to investigate the potential anticancer activity and underlying mechanisms of rhein in oral cancer cells. The antigrowth effect of rhein in oral cancer cells was estimated by cell proliferation, soft agar colony formation, migration, and invasion assay. The cell cycle and apoptosis were detected by flow cytometry. The underlying mechanism of rhein in oral cancer cells was explored by immunoblotting. The in vivo anticancer effect was evaluated by oral cancer xenografts. Rhein significantly inhibited oral cancer cell growth by inducing apoptosis and S-phase cell cycle arrest. Rhein inhibited oral cancer cell migration and invasion through the regulation of epithelial-mesenchymal transition-related proteins. Rhein induced reactive oxygen species (ROS) accumulation in oral cancer cells to inhibit the AKT/mTOR signaling pathway. Rhein exerted anticancer activity in vitro and in vivo by inducing oral cancer cell apoptosis and ROS via the AKT/mTOR signaling pathway in oral cancer. Rhein is a potential therapeutic drug for oral cancer treatment.
Collapse
Affiliation(s)
- Haibo Zhang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Lei Ma
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Eungyung Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Junkoo Yi
- School of Animal Life Convergence Science, Hankyung National University, Anseong 17579, Republic of Korea
| | - Hai Huang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Hyeonjin Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Muhammad Atif Raza
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Sijun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Soyoung Jang
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kirim Kim
- Department of Dental Hygiene, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Sung-Hyun Kim
- Department of Bio-Medical Analysis, Korea Polytechnic College, Chungnam 34134, Republic of Korea
| | - Youngkyun Lee
- School of Dentistry, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eunkyong Kim
- Department of Dental Hygiene, Kyungpook National University, Sangju 37224, Republic of Korea
| | - Zae Young Ryoo
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea
- Department of Animal Biotechnology, Research Center for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea
| |
Collapse
|
11
|
Zhou C, Gong T, Chen J, Chen T, Yang J, Zhu P. Production of a Novel Protopanaxatriol-Type Ginsenoside by Yeast Cell Factories. Bioengineering (Basel) 2023; 10:bioengineering10040463. [PMID: 37106650 PMCID: PMC10135449 DOI: 10.3390/bioengineering10040463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/24/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023] Open
Abstract
Ginsenosides, the main active compounds in Panax species, are glycosides of protopanaxadiol (PPD) or protopanaxatriol (PPT). PPT-type ginsenosides have unique pharmacological activities on the central nervous system and cardiovascular system. As an unnatural ginsenoside, 3,12-Di-O-β-D-glucopyranosyl-dammar-24-ene-3β,6α,12β,20S-tetraol (3β,12β-Di-O-Glc-PPT) can be synthesized through enzymatic reactions but is limited by the expensive substrates and low catalytic efficiency. In the present study, we successfully produced 3β,12β-Di-O-Glc-PPT in Saccharomyces cerevisiae with a titer of 7.0 mg/L by expressing protopanaxatriol synthase (PPTS) from Panax ginseng and UGT109A1 from Bacillus subtilis in PPD-producing yeast. Then, we modified this engineered strain by replacing UGT109A1 with its mutant UGT109A1-K73A, overexpressing the cytochrome P450 reductase ATR2 from Arabidopsis thaliana and the key enzymes of UDP-glucose biosynthesis to increase the production of 3β,12β-Di-O-Glc-PPT, although these strategies did not show any positive effect on the yield of 3β,12β-Di-O-Glc-PPT. However, the unnatural ginsenoside 3β,12β-Di-O-Glc-PPT was produced in this study by constructing its biosynthetic pathway in yeast. To the best of our knowledge, this is the first report of producing 3β,12β-Di-O-Glc-PPT through yeast cell factories. Our work provides a viable route for the production of 3β,12β-Di-O-Glc-PPT, which lays a foundation for drug research and development.
Collapse
Affiliation(s)
- Chen Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Ting Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jingjing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Tianjiao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jinling Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Ping Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC Key Laboratory of Biosynthesis of Natural Products, CAMS Key Laboratory of Enzyme and Biocatalysis of Natural Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
12
|
The ways for ginsenoside Rh2 to fight against cancer: the molecular evidences in vitro and in vivo. J Ginseng Res 2023; 47:173-182. [PMID: 36926617 PMCID: PMC10014223 DOI: 10.1016/j.jgr.2022.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/30/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer is a global public health issue that becomes the second primary cause of death globally. Considering the side effects of radio- or chemo-therapy, natural phytochemicals are promising alternatives for therapeutic interventions to alleviate the side effects and complications. Ginsenoside Rh2 (GRh2) is the main phytochemical extracted from Panax ginseng C.A. Meyer with anticancer activity. GRh2 could induce apoptosis and autophagy of cancer cells and inhibit proliferation, metastasis, invasion, and angiogenesis in vitro and in vivo. In addition, GRh2 could be used as an adjuvant to chemotherapeutics to enhance the anticancer effect and reverse the adverse effects. Here we summarized the understanding of the molecular mechanisms underlying the anticancer effects of GRh2 and proposed future directions to promote the development and application of GRh2.
Collapse
|
13
|
Jung DH, Nahar J, Mathiyalagan R, Rupa EJ, Ramadhania ZM, Han Y, Yang DC, Kang SC. A Focused Review on Molecular Signalling Mechanisms of Ginsenosides Anti-Lung Cancer and Anti-inflammatory Activities. Anticancer Agents Med Chem 2023; 23:3-14. [PMID: 35319393 DOI: 10.2174/1871520622666220321091022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/01/2021] [Accepted: 12/12/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ginseng (Panax ginseng Meyer) is a cultivated medicinal herb that has been widely available in the Asian region since the last century. Ginseng root is used worldwide in Oriental medicine. Currently, the global mortality and infection rates for lung cancer and inflammation are significantly increasing. Therefore, various preventative methods related to the activity of ginsenosides have been used for lung cancer as well as inflammation. METHODS Web-based searches were performed on Web of Science, Springer, PubMed, and Scopus. A cancer statistical analysis was also conducted to show the current ratio of affected cases and death from lung cancer around the world. RESULTS Ginsenosides regulate the enzymes that participate in tumor growth and migration, such as nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (p38 MAPK), c-Jun N-terminal kinase (JNK), extracellular signalregulated kinases 1/2 (ERK1/2), the gelatinase network metalloproteinase-2 (MMP-2/9) and activator protein 1 (AP-1). In addition, ginsenosides also possess anti-inflammatory effects by inhibiting the formation of proinflammatory cytokines (tumor necrosis factor-α) (TNF-α) and interleukin-1β (IL-1β) and controlling the activities of inflammatory signalling pathways, such as NF-κB, Janus kinase2/signal transducer, and activator of transcription 3 (Jak2/Stat3). CONCLUSION In several in vitro and in vivo models, P. ginseng showed potential beneficial effects in lung cancer and inflammation treatment. In this review, we provide a detailed and up-to-date summary of research evidence for antilung cancer and anti-inflammatory protective effects of ginsenosides and their potential molecular mechanisms.
Collapse
Affiliation(s)
- Dae-Hyo Jung
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Jinnatun Nahar
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Esrat Jahan Rupa
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Zelika Mega Ramadhania
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Yaxi Han
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Deok-Chun Yang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.,Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Se Chan Kang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| |
Collapse
|
14
|
Recent advances in ginsenosides against respiratory diseases: Therapeutic targets and potential mechanisms. Biomed Pharmacother 2023; 158:114096. [PMID: 36502752 DOI: 10.1016/j.biopha.2022.114096] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Respiratory diseases mainly include asthma, influenza, pneumonia, chronic obstructive pulmonary disease, pulmonary hypertension, lung fibrosis, and lung cancer. Given their high prevalence and poor prognosis, the prevention and treatment of respiratory diseases are increasingly essential. In particular, the development for the novel strategies of drug treatment has been a hot topic in the research field. Ginsenosides are the major component of Panax ginseng C. A. Meyer (ginseng), a food homology and well-known medicinal herb. In this review, we summarize the current therapeutic effects and molecular mechanisms of ginsenosides in respiratory diseases. METHODS The reviewed studies were retrieved via a thorough analysis of numerous articles using electronic search tools including Sci-Finder, ScienceDirect, PubMed, and Web of Science. The following keywords were used for the online search: ginsenosides, asthma, influenza, pneumonia, chronic obstructive pulmonary disease (COPD), pulmonary hypertension (PH), lung fibrosis, lung cancer, and clinical trials. We summarized the findings and the conclusions from 176 manuscripts on ginsenosides, including research articles and reviews. RESULTS Ginsenosides Rb1, Rg1, Rg3, Rh2, and CK, which are the most commonly reported ginsenosides for treating of respiratory diseases, and other ginsenosides such as Rh1, Rk1, Rg5, Rd and Re, all primarily reduce pneumonia, fibrosis, and inhibit tumor progression by targeting NF-κB, TGF-β/Smad, PI3K/AKT/mTOR, and JNK pathways, thereby ameliorating respiratory diseases. CONCLUSION This review provides novel ideas and important aspects for the future research of ginsenosides for treating respiratory diseases.
Collapse
|
15
|
Li J, Huang T, Lu J, Xu X, Zhang W. Metabonomic profiling of clubroot-susceptible and clubroot-resistant radish and the assessment of disease-resistant metabolites. FRONTIERS IN PLANT SCIENCE 2022; 13:1037633. [PMID: 36570889 PMCID: PMC9772615 DOI: 10.3389/fpls.2022.1037633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Plasmodiophora brassicae causes a serious threat to cruciferous plants including radish (Raphanus sativus L.). Knowledge on the pathogenic regularity and molecular mechanism of P. brassicae and radish is limited, especially on the metabolism level. In the present study, clubroot-susceptible and clubroot-resistant cultivars were inoculated with P. brassicae Race 4, root hairs initial infection of resting spores (107 CFU/mL) at 24 h post-inoculation and root galls symptom arising at cortex splitting stage were identified on both cultivars. Root samples of cortex splitting stage of two cultivars were collected and used for untargeted metabonomic analysis. We demonstrated changes in metabolite regulation and pathways during the cortex splitting stage of diseased roots between clubroot-susceptible and clubroot-resistant cultivars using untargeted metabonomic analysis. We identified a larger number of differentially regulated metabolites and heavier metabolite profile changes in the susceptible cultivar than in the resistant counterpart. The metabolites that were differentially regulated in both cultivars were mostly lipids and lipid-like molecules. Significantly regulated metabolites and pathways according to the P value and variable important in projection score were identified. Moreover, four compounds, including ethyl α-D-thioglucopyranoside, imipenem, ginsenoside Rg1, and 6-gingerol, were selected, and their anti-P. brassicae ability and effects on seedling growth were verified on the susceptible cultivar. Except for ethyl α-D-thioglucopyranoside, the remaining could inhibit clubroot development of varing degree. The use of 5 mg/L ginsenoside Rg1 + 5 mg/L 6-gingerol resulted in the lowest disease incidence and disease index among all treatments and enhanced seedling growth. The regulation of pathways or metabolites of carbapenem and ginsenoside was further explored. The results provide a preliminary understanding of the interaction between radish and P. brassicae at the metabolism level, as well as the development of measures for preventing clubroot.
Collapse
Affiliation(s)
- Jingwei Li
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Tingmin Huang
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Jinbiao Lu
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Xiuhong Xu
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| | - Wanping Zhang
- Vegetable Research Institute, Guizhou University, Guiyang, China
- College of Agriculture, Guizhou University, Guiyang, China
| |
Collapse
|
16
|
Role of ginsenoside Rh2 in tumor therapy and tumor microenvironment immunomodulation. Biomed Pharmacother 2022; 156:113912. [DOI: 10.1016/j.biopha.2022.113912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/24/2022] Open
|
17
|
Ginsenosides in cancer: A focus on the regulation of cell metabolism. Biomed Pharmacother 2022; 156:113756. [DOI: 10.1016/j.biopha.2022.113756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/17/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
|
18
|
Abstract
As a steroid skeleton-based saponin, ginsenoside Rh2 (G-Rh2) is one of the major bioactive ginsenosides from the plants of genus Panax L. Many studies have reported the notable pharmacological activities of G-Rh2 such as anticancer, antiinflammatory, antiviral, antiallergic, antidiabetic, and anti-Alzheimer's activities. Numerous preclinical studies have demonstrated the great potential of G-Rh2 in the treatment of a wide range of carcinomatous diseases in vitro and in vivo. G-Rh2 is able to inhibit proliferation, induce apoptosis and cell cycle arrest, retard metastasis, promote differentiation, enhance chemotherapy and reverse multi-drug resistance against multiple tumor cells. The present review mainly summarizes the anticancer effects and related mechanisms of G-Rh2 in various models as well as the recent advances in G-Rh2 delivery systems and structural modification to ameliorate its anticancer activity and pharmacokinetics characteristics.
Collapse
|
19
|
Ginsenoside Rh2 Induces HeLa Apoptosis through Upregulating Endoplasmic Reticulum Stress-Related and Downstream Apoptotic Gene Expression. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227865. [PMID: 36431966 PMCID: PMC9699401 DOI: 10.3390/molecules27227865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Cervical cancer is a common gynecological malignancy afflicting women all over the world. Ginsenoside Rh2 (GRh2), especially 20(S)-GRh2, is a biologically active component in the natural plant ginseng, which can exhibit anticancer effects. Here, we aimed to investigate the effect of 20(S)-GRh2 on cervical cancer and elucidate the underlying mechanism through RNA-seq. In this study, the CCK-8 assay showed that 20(S)-GRh2 inhibited HeLa cell viability in a time- and dose-dependent manner. Caspase 3 activity and Annexin V staining results showed that 20(S)-GRh2 induced apoptosis of HeLa cells. Gene function enrichment analysis revealed that the biological process gene ontology (GO) terms were associated with the apoptotic signaling pathway. Biological process GO terms' similarity network indicated that apoptosis might be from endoplasmic reticulum stress (ERs). Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that 20(S)-GRh2 primarily modulates apoptosis pathway genes. Combined protein-protein interaction network, hub gene screening, and qPCR validation data showed that ERs-related genes (ATF4 and DDIT3) and the downstream apoptotic genes (JUN, FOS, BBC3, and PMAIP1) were potential novel targets of 20(S)-GRh2-inducing cervical cancer cell apoptosis. Differential transcript usage analysis indicated that DDIT3 is also a differential transcript and its usage of the isoform (ENST00000552740.5) was reduced by 20(S)-GRh2. Molecular docking suggested that 20(S)-GRh2 binds to the targets (ATF4, DDIT3, JUN, FOS, BBC3, and PMAIP1) with high affinity. In conclusion, our findings indicated that 20(S)-GRh2 might promote ERs-related apoptosis of cervical cancer cells by regulating the DDIT3-based targets' signal pathway. The role of 20(S)-GRh2 at the transcriptome level provides novel targets and evidence for the treatment of cervical cancer.
Collapse
|
20
|
Chen H, Cai Y, Sun S, Pan Z, Han Z, Liu P, Liu Y. Repair effect of photobiomodulation combined with human umbilical cord mesenchymal stem cells on rats with acute lung injury. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 234:112541. [PMID: 36029758 DOI: 10.1016/j.jphotobiol.2022.112541] [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: 07/05/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
Acute lung injury (ALI) impaired the function of blood oxygen exchange function, resulting in tissue hypoxia and patient death. Recently, human umbilical cord mesenchymal stem cells (hUCMSCs) are thought to mitigate the effects of ALI, which boosts researchers' interest in employing stem cell-based therapies to manage ALI. However, as a novel therapy, hUCMSCs still face various limitations such as migrating weakly and insufficient proliferation in vivo. Photobiomodulation (PBM) effciently promotes cell proliferation, migration and homing, which presents a promising strategy for overcoming above limitations. In this study, PBM was emerged to intervene hUCMSCs through detecting cell proliferation, oxidative stress-related factors and inflammatory factors. These results assuredly confirmed that PBM enhanced the antioxidant capacity of cells and improved cell survival in vitro experiments. In vivo, PBM-intervened hUCMSCs intuitively reduce thickness of alveolar septum, excessive secretion of inflammatory factors, relieves bleeding, edema and fibrosis. As a physical intervention, PBM further strengthens the therapeutic effect of hUCMSCs and depicted a hopeful therapy in ALI treatment.
Collapse
Affiliation(s)
- Hongli Chen
- State Key Laboratry of Separation Membrane and Membrane Process, Tiangong University, Tianjin 300387, China; Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin 300457, China
| | - Yuanhao Cai
- State Key Laboratry of Separation Membrane and Membrane Process, Tiangong University, Tianjin 300387, China
| | - Shujie Sun
- State Key Laboratry of Separation Membrane and Membrane Process, Tiangong University, Tianjin 300387, China
| | - Zhenhua Pan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhibo Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin 300457, China
| | - Pai Liu
- State Key Laboratry of Separation Membrane and Membrane Process, Tiangong University, Tianjin 300387, China.
| | - Yi Liu
- State Key Laboratry of Separation Membrane and Membrane Process, Tiangong University, Tianjin 300387, China.
| |
Collapse
|
21
|
Zhu X, Chen X, Wang G, Lei D, Chen X, Lin K, Li M, Lin H, Li D, Zheng Q. Picropodophyllin Inhibits the Proliferation of Human Prostate Cancer DU145 and LNCaP Cells <i>via</i> ROS Production and PI3K/AKT Pathway Inhibition. Biol Pharm Bull 2022; 45:1027-1035. [DOI: 10.1248/bpb.b21-01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Xuejie Zhu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Xiaojie Chen
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Guoli Wang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Dan Lei
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Xiaoyu Chen
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Kehao Lin
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Minjing Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Haiyan Lin
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Defang Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| | - Qiusheng Zheng
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University
| |
Collapse
|
22
|
An mTOR and DNA-PK dual inhibitor CC-115 hinders non-small cell lung cancer cell growth. Cell Death Dis 2022; 8:293. [PMID: 35717530 PMCID: PMC9206683 DOI: 10.1038/s41420-022-01082-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022]
Abstract
Molecularly-targeted agents are still urgently needed for better non-small cell lung cancer (NSCLC) therapy. CC-115 is a potent DNA-dependent protein kinase (DNA-PK) and mammalian target of rapamycin (mTOR) dual blocker. We evaluated its activity in different human NSCLC cells. In various primary human NSCLC cells and A549 cells, CC-115 potently inhibited viability, cell proliferation, cell cycle progression, and hindered cell migration/invasion. Apoptosis was provoked in CC-115-stimulated NSCLC cells. The dual inhibitor, however, was unable to induce significant cytotoxic and pro-apoptotic activity in the lung epithelial cells. In primary NSCLC cells, CC-115 blocked activation of mTORC1/2 and DNA-PK. Yet, CC-115-induced primary NSCLC cell death was more potent than combined inhibition of DNA-PK plus mTOR. Further studies found that CC-115 provoked robust oxidative injury in primary NSCLC cells, which appeared independent of mTOR-DNA-PK dual blockage. In vivo studies showed that CC-115 oral administration in nude mice remarkably suppressed primary NSCLC cell xenograft growth. In CC-115-treated NSCLC xenograft tissues, mTOR-DNA-PK dual inhibition and oxidative injury were detected. Together, CC-115 potently inhibits NSCLC cell growth.
Collapse
|
23
|
Song C, Yuan Y, Zhou J, He Z, Hu Y, Xie Y, Liu N, Wu L, Zhang J. Network Pharmacology-Based Prediction and Verification of Ginsenoside Rh2-Induced Apoptosis of A549 Cells via the PI3K/Akt Pathway. Front Pharmacol 2022; 13:878937. [PMID: 35600856 PMCID: PMC9114502 DOI: 10.3389/fphar.2022.878937] [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: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
Ginsenoside Rh2 (G-Rh2), a rare protopanaxadiol (PPD)-type triterpene saponin, from Panax ginseng has anti-proliferation, anti-invasion, and anti-metastatic activity. However, the mechanisms by which G-Rh2 induces apoptosis of lung cancer cells are unclear. In the present work, a G-Rh2 target-lung cancer network was constructed and analyzed by the network pharmacology approach. A total of 91 compound-targets of G-Rh2 was obtained based on the compound-target network analysis, and 217 targets were identified for G-Rh2 against lung cancer by PPI network analysis. The 217 targets were significantly enriched in 103 GO terms with FDR <0.05 as threshold in the GO enrichment analysis. In KEGG pathway enrichment analysis, all the candidate targets were significantly enriched in 143 pathways, among of which PI3K-Akt signaling pathway was identified as one of the top enriched pathway. Besides, G-Rh2 induced apoptosis in human lung epithelial (A549) cells was verified in this work. G-Rh2 significantly inhibited the proliferation of A549 cells in a dose-dependent manner, and the apoptosis rate significantly increased from 4.4% to 78.7% using flow cytometry. Western blot analysis revealed that the phosphorylation levels of p85, PDK1, Akt and IκBα were significantly suppressed by G-Rh2. All the experimental findings were consistent with the network pharmacology results. Research findings in this work will provide potential therapeutic value for further mechanism investigations.
Collapse
Affiliation(s)
- Chao Song
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Sciences, Huaiyin Normal University, Huaian, China
| | - Yue Yuan
- School of Pharmaceutical Sciences, Institute for Chinese Materia Medica, Tsinghua University, Beijing, China
| | - Jing Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Sciences, Huaiyin Normal University, Huaian, China
| | - Ziliang He
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Sciences, Huaiyin Normal University, Huaian, China
| | - Yeye Hu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Sciences, Huaiyin Normal University, Huaian, China
| | - Yuan Xie
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Sciences, Huaiyin Normal University, Huaian, China
| | - Nan Liu
- Beijing Increasepharm Safety and Efficacy Co., Ltd, Beijing, China
- *Correspondence: Nan Liu, ; Lei Wu, ; Ji Zhang,
| | - Lei Wu
- Institute of Applied Chemistry, Academy of Sciences, Nanchang, China
- *Correspondence: Nan Liu, ; Lei Wu, ; Ji Zhang,
| | - Ji Zhang
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture and Environmental Protection, School of Life Sciences, Huaiyin Normal University, Huaian, China
- *Correspondence: Nan Liu, ; Lei Wu, ; Ji Zhang,
| |
Collapse
|
24
|
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
|
25
|
Guo S, Li Y, Su H, Meng M, Xi J, Mo G, Chen X. Aidi injection as adjunctive treatment to gemcitabine-based chemotherapy for advanced non-small cell lung cancer: a systematic review and meta-analysis. PHARMACEUTICAL BIOLOGY 2021; 59:1260-1275. [PMID: 34541998 PMCID: PMC8451693 DOI: 10.1080/13880209.2021.1973038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/05/2021] [Accepted: 08/21/2021] [Indexed: 05/29/2023]
Abstract
CONTEXT Aidi injection is one of the most commonly use antitumor Chinese medicine injections for advanced non-small cell lung cancer (NSCLC). It is made from the extraction of Astragalus, Eleutherococcus senticosus, Ginseng, and Cantharis. OBJECTIVE To evaluate the efficacy and safety of Aidi injection in combination with gemcitabine-based chemotherapy (GBC) for advanced NSCLC. MATERIALS AND METHODS PubMed, Embase, Cochrane Library, Chinese Biological Medicine, China National Knowledge Infrastructure, Wanfang, and VIP were searched for relevant randomised controlled trials (RCTs) comparing Aidi injection plus GBC treatment with GBC alone in NSCLC, from inception up to October 2020. The primary outcomes were objective response rate (ORR), and disease control rate (DCR). Secondary outcomes were quality of life (QOL) and adverse drug reactions (ADRs). The quality of evidence was rated using the GRADE approach. This study was registered with PROSPERO: CRD42021221225. RESULTS In total, 54 RCTs involving 4318 NSCLC patients were included in this meta-analysis. Compared with GBC alone, Aidi injection plus GBC significantly improve ORR (risk ratios [RR] = 1.38, 95% confidence interval [CI] 1.29-1.48), DCR (RR = 1.15, 95% CI 1.12-1.19), QOL (RR = 1.71, 95% CI 1.54-1.89), and reduced the risk of gastrointestinal toxicity, thrombocytopenia, neutropenia, liver injury, renal injury, and anaemia. The evaluation results of the evidence ranged from moderate to low. CONCLUSIONS Current moderate evidence revealed that Aidi injection as an adjunctive treatment to GBC was associated with superior benefits in patients with advanced NSCLC and alleviate toxicities. High-quality RCTs are needed to further confirm the results.
Collapse
Affiliation(s)
- Sitong Guo
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yan Li
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Henghai Su
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Mingyu Meng
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jiaxi Xi
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Guangyan Mo
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Xiaoyu Chen
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| |
Collapse
|
26
|
Li X, Chen M, Yao Z, Du H, Zhang T, Wang H, Xie Y, Li Z. Jujuboside B induces mitochondrial-dependent apoptosis in colorectal cancer through ROS-mediated PI3K/Akt pathway in vitro and in vivo. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
27
|
Xu C, Li L, Wang C, Jiang J, Li L, Zhu L, Jin S, Jin Z, Lee JJ, Li G, Yan G. Effects of G-Rh2 on mast cell-mediated anaphylaxis via AKT-Nrf2/NF-κB and MAPK-Nrf2/NF-κB pathways. J Ginseng Res 2021; 46:550-560. [PMID: 35818417 PMCID: PMC9270651 DOI: 10.1016/j.jgr.2021.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/04/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022] Open
Abstract
Background The effect of ginsenoside Rh2 (G-Rh2) on mast cell-mediated anaphylaxis remains unclear. Herein, we investigated the effects of G-Rh2 on OVA-induced asthmatic mice and on mast cell-mediated anaphylaxis. Methods Asthma model was established for evaluating airway changes and ear allergy. RPMCs and RBL-2H3 were used for in vitro experiments. Calcium uptake, histamine release and degranulation were detected. ELISA and Western blot measured cytokine and protein levels, respectively. Results G-Rh2 inhibited OVA-induced airway remodeling, the production of TNF-α, IL-4, IL-8, IL-1β and the degranulation of mast cells of asthmatic mice. G-Rh2 inhibited the activation of Syk and Lyn in lung tissue of OVA-induced asthmatic mice. G-Rh2 inhibited serum IgE production in OVA induced asthmatic mice. Furthermore, G-Rh2 reduced the ear allergy in IgE-sensitized mice. G-Rh2 decreased the ear thickness. In vitro experiments G-Rh2 significantly reduced calcium uptake and inhibited histamine release and degranulation in RPMCs. In addition, G-Rh2 reduced the production of IL-1β, TNF-α, IL-8, and IL-4 in IgE-sensitized RBL-2H3 cells. Interestingly, G-Rh2 was involved in the FcεRI pathway activation of mast cells and the transduction of the Lyn/Syk signaling pathway. G-Rh2 inhibited PI3K activity in a dose-dependent manner. By blocking the antigen-induced phosphorylation of Lyn, Syk, LAT, PLCγ2, PI3K ERK1/2 and Raf-1 expression, G-Rh2 inhibited the NF-κB, AKT-Nrf2, and p38MAPK-Nrf2 pathways. However, G-Rh2 up-regulated Keap-1 expression. Meanwhile, G-Rh2 reduced the levels of p-AKT, p38MAPK and Nrf2 in RBL-2H3 sensitized IgE cells and inhibited NF-κB signaling pathway activation by activating the AKT-Nrf2 and p38MAPK-Nrf2 pathways. Conclusion G-Rh2 inhibits mast cell-induced allergic inflammation, which might be mediated by the AKT-Nrf2/NF-κB and p38MAPK-Nrf2/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Chang Xu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Jingzhi Jiang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
| | - Lianhua Zhu
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Department of Dermatology, Yanbian University Hospital, Yanji, China
| | - Shan Jin
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Department of Dermatology, Yanbian University Hospital, Yanji, China
| | - Zhehu Jin
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Department of Dermatology, Yanbian University Hospital, Yanji, China
| | - Jung Joon Lee
- College of Pharmacy, Yanbian University, Yanji, China
| | - Guanhao Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Food Research Center of Yanbian University, Yanji, China
- Corresponding author. Food Research Center of Yanbian University, No. 977 Gongyuan Road, Yanji, 133002, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji, China
- Corresponding author. Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, No. 977 Gongyuan Road, Yanji, 133002, PR China.
| |
Collapse
|
28
|
Liu L, Wang H, Chai X, Meng Q, Jiang S, Zhao F. Advances in Biocatalytic Synthesis, Pharmacological Activities, Pharmaceutical Preparation and Metabolism of Ginsenoside Rh2. Mini Rev Med Chem 2021; 22:437-448. [PMID: 34517798 DOI: 10.2174/1389557521666210913114631] [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: 10/30/2020] [Revised: 01/25/2021] [Accepted: 06/22/2021] [Indexed: 11/22/2022]
Abstract
Ginsenoside Rh2 (3β-O-Glc-protopanaxadiol), a trace but characteristic pharmacological component of red ginseng, exhibited versatile pharmacological activities, such as antitumor effects, improved cardiac function and fibrosis, anti-inflammatory effects, antibiosis and excellent medicinal potential. In recent years, increased research has been performed on the biocatalytic synthesis of ginsenoside Rh2. In this paper, advances in the biocatalytic synthesis, pharmacological activities, pharmaceutical preparation and metabolism of ginsenoside Rh2 are reviewed.
Collapse
Affiliation(s)
- Li Liu
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005. China
| | - Huiyun Wang
- College of Pharmacy, Jining Medical University, Shandong Province, 276826. China
| | - Xiaoyun Chai
- School of Pharmacy, Naval Medical University, Shanghai, 200433. China
| | - Qingguo Meng
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005. China
| | - Sheng Jiang
- Shandong Wendeng Jizhen American Ginseng Industry Co., Ltd., Shandong Province, 264400. China
| | - Fenglan Zhao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005. China
| |
Collapse
|
29
|
Anti-Cancer Effect of Panax Ginseng and Its Metabolites: From Traditional Medicine to Modern Drug Discovery. Processes (Basel) 2021. [DOI: 10.3390/pr9081344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer incidence and mortality rate are growing worldwide. The effectiveness of cancer therapy depends on the degree of cancer development. Anticancer prevention, screening tests, detection of precancerous conditions or cancers at an early stage of development help to prevent the development of cancer, and in the event of cancer development, they provide the best chance for a full recovery. However, in most cases of advanced cancer, there is no method that can fully cure this disease. Recently, natural products have gained more attention in cancer therapy. Panax ginseng (PG), one of the most popular natural products, is reported to have a wide range of pharmacological activities in cancer. Therefore, the anti-cancer effects and mechanisms of PG and its metabolites (compound K, Ginsenoside Rh1, Rh2, Rh3 and F1) in five major cancers (lung cancer, breast cancer, colon cancer, prostate cancer and stomach cancer) are reviewed in this study. It is confirmed that PG and its metabolites regulated apoptosis, epithelial mesenchymal transition (EMT), angiogenesis, cell cycle arrest and multidrug resistance (MDR) in vitro and in vivo cancer models. In particular, ginsenoside Rh2 showed anticancer effects in all five major cancers. This review could improve the understanding of anticancer mechanisms of PG and its metabolites against major five cancers. Further clinical studies are needed for development anti-cancer drugs using PG and its metabolites.
Collapse
|
30
|
Zhang H, Yi JK, Huang H, Park S, Kwon W, Kim E, Jang S, Kim SY, Choi SK, Yoon D, Kim SH, Liu K, Dong Z, Ryoo ZY, Kim MO. 20 (S)-ginsenoside Rh2 inhibits colorectal cancer cell growth by suppressing the Axl signaling pathway in vitro and in vivo. J Ginseng Res 2021; 46:396-407. [PMID: 35600769 PMCID: PMC9120647 DOI: 10.1016/j.jgr.2021.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/20/2022] Open
Abstract
Background Colorectal cancer (CRC) has a high morbidity and mortality worldwide. 20 (S)-ginsenoside Rh2 (G-Rh2) is a natural compound extracted from ginseng, which exhibits anticancer effects in many cancer types. In this study, we demonstrated the effect and underlying molecular mechanism of G-Rh2 in CRC cells in vitro and in vivo. Methods Cell proliferation, migration, invasion, apoptosis, cell cycle, and western blot assays were performed to evaluate the effect of G-Rh2 on CRC cells. In vitro pull-down assay was used to verify the interaction between G-Rh2 and Axl. Transfection and infection experiments were used to explore the function of Axl in CRC cells. CRC xenograft models were used to further investigate the effect of Axl knockdown and G-Rh2 on tumor growth in vivo. Results G-Rh2 significantly inhibited proliferation, migration, and invasion, and induced apoptosis and G0/G1 phase cell cycle arrest in CRC cell lines. G-Rh2 directly binds to Axl and inhibits the Axl signaling pathway in CRC cells. Knockdown of Axl suppressed the growth, migration and invasion ability of CRC cells in vitro and xenograft tumor growth in vivo, whereas overexpression of Axl promoted the growth, migration, and invasion ability of CRC cells. Moreover, G-Rh2 significantly suppressed CRC xenograft tumor growth by inhibiting Axl signaling with no obvious toxicity to nude mice. Conclusion Our results indicate that G-Rh2 exerts anticancer activity in vitro and in vivo by suppressing the Axl signaling pathway. G-Rh2 is a promising candidate for CRC prevention and treatment.
Collapse
Affiliation(s)
- Haibo Zhang
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju, Republic of Korea
| | - Jun-Koo Yi
- Gyeongbuk Livestock Research Institute, Yeongju, Republic of Korea
| | - Hai Huang
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju, Republic of Korea
| | - Sijun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, Republic of Korea
| | - Wookbong Kwon
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
| | - Eungyung Kim
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju, Republic of Korea
| | - Soyoung Jang
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, Republic of Korea
| | - Si-Yong Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, Republic of Korea
| | - Seong-kyoon Choi
- Division of Biotechnology, DGIST, Daegu, Republic of Korea
- Core Protein Resources Center, DGIST, Daegu, Republic of Korea
| | - Duhak Yoon
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju, Republic of Korea
| | - Sung-Hyun Kim
- Department of Bio-Medical Analysis, Korea Polytechnic College, Chungnam, Republic of Korea
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Zae Young Ryoo
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch, Kyungpook National University, Daegu, Republic of Korea
- Corresponding author.
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju, Republic of Korea
- Corresponding author. Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju, Gyeongsangbukdo, 37224, Republic of Korea.
| |
Collapse
|
31
|
Zha JH, Xia YC, Ye CL, Hu Z, Zhang Q, Xiao H, Yu BT, Xu WH, Xu GQ. The Anti-Non-Small Cell Lung Cancer Cell Activity by a mTOR Kinase Inhibitor PQR620. Front Oncol 2021; 11:669518. [PMID: 34178653 PMCID: PMC8222575 DOI: 10.3389/fonc.2021.669518] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/11/2021] [Indexed: 11/21/2022] Open
Abstract
In non-small-cell lung carcinoma (NSCLC), aberrant activation of mammalian target of rapamycin (mTOR) contributes to tumorigenesis and cancer progression. PQR620 is a novel and highly-potent mTOR kinase inhibitor. We here tested its potential activity in NSCLC cells. In primary human NSCLC cells and established cell lines (A549 and NCI-H1944), PQR620 inhibited cell growth, proliferation, and cell cycle progression, as well as cell migration and invasion, while inducing significant apoptosis activation. PQR620 disrupted assembles of mTOR complex 1 (mTOR-Raptor) and mTOR complex 2 (mTOR-Rictor-Sin1), and blocked Akt, S6K1, and S6 phosphorylations in NSCLC cells. Restoring Akt-mTOR activation by a constitutively-active Akt1 (S473D) only partially inhibited PQR620-induced cytotoxicity in NSCLC cells. PQR620 was yet cytotoxic in Akt1/2-silenced NSCLC cells, supporting the existence of Akt-mTOR-independent mechanisms. Indeed, PQR620 induced sphingosine kinase 1 (SphK1) inhibition, ceramide production and oxidative stress in primary NSCLC cells. In vivo studies demonstrated that daily oral administration of a single dose of PQR620 potently inhibited primary NSCLC xenograft growth in severe combined immune deficient mice. In PQR620-treated xenograft tissues, Akt-mTOR inactivation, apoptosis induction, SphK1 inhibition and oxidative stress were detected. In conclusion, PQR620 exerted potent anti-NSCLC cell activity via mTOR-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Jian-Hua Zha
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying-Chen Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chun-Lin Ye
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhi Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qin Zhang
- Department of Respiratory Medicine, Suzhou Hospital Affiliated Nanjing Medical University, Suzhou, China
| | - Han Xiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei-Hua Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Guo-Qiu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
32
|
Paving the Road Toward Exploiting the Therapeutic Effects of Ginsenosides: An Emphasis on Autophagy and Endoplasmic Reticulum Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:137-160. [PMID: 33861443 DOI: 10.1007/978-3-030-64872-5_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Programmed cell death processes such as apoptosis and autophagy strongly contribute to the onset and progression of cancer. Along with these lines, modulation of cell death mechanisms to combat cancer cells and elimination of resistance to apoptosis is of great interest. It appears that modulation of autophagy and endoplasmic reticulum (ER) stress with specific agents would be beneficial in the treatment of several disorders. Interestingly, it has been suggested that herbal natural products may be suitable candidates for the modulation of these processes due to few side effects and significant therapeutic potential. Ginsenosides are derivatives of ginseng and exert modulatory effects on the molecular mechanisms associated with autophagy and ER stress. Ginsenosides act as smart phytochemicals that confer their effects by up-regulating ATG proteins and converting LC3-I to -II, which results in maturation of autophagosomes. Not only do ginsenosides promote autophagy but they also possess protective and therapeutic properties due to their capacity to modulate ER stress and up- and down-regulate and/or dephosphorylate UPR transducers such as IRE1, PERK, and ATF6. Thus, it would appear that ginsenosides are promising agents to potentially restore tissue malfunction and possibly eliminate cancer.
Collapse
|
33
|
Wei XM, Jiang S, Li SS, Sun YS, Wang SH, Liu WC, Wang Z, Wang YP, Zhang R, Li W. Endoplasmic Reticulum Stress-Activated PERK-eIF2α-ATF4 Signaling Pathway is Involved in the Ameliorative Effects of Ginseng Polysaccharides against Cisplatin-Induced Nephrotoxicity in Mice. ACS OMEGA 2021; 6:8958-8966. [PMID: 33842766 PMCID: PMC8027996 DOI: 10.1021/acsomega.0c06339] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 05/25/2023]
Abstract
Although previous studies have reported that saponins (ginsenosides, the major active and most representative ingredients in Panax ginseng C.A. Meyer) exerted a good ameliorative effect on cisplatin (CP)-induced acute kidney injury in animal models, little attention has been paid to a large number of polysaccharides isolated and purified from ginseng. This work aimed to investigate the protective effect and the possible molecular mechanism of ginseng polysaccharide (WGP) on CP-induced kidney toxicology in mice. The results from biomarker analysis including serum creatinine (CRE) and blood urea nitrogen (BUN) confirmed the protective effect of WGP at 200 and 400 mg/kg on CP-induced renal-toxicology. We found that WGP reduces the apoptosis of kidney cells by inhibiting endoplasmic reticulum (ER) stress caused by CP, which is manifested by increased phosphorylation of PERK. In addition, the apoptosis-associated with caspase 3 activation in renal cells induced by CP was inhibited after administration of WGP, and the phosphorylation levels of PI3K and AKT were also reduced significantly. We also demonstrated that after exposure to CP, the unfolded protein response signaling pathway PERK-eIF2α-ATF4 axis was significantly activated, manifested by increased phosphorylation of eIF2α and increased expression of ATF4 and CHOP. Interestingly, the WGP administration improves this situation. Furthermore, the supplement of WGP inhibited the overexpression of nuclear factor-kappa B p65 (NF-κB p65) and tumor necrosis factor-α (TNF-α) caused by CP exposure. In short, for the first time, our findings indicated that WGP could effectively prevent CP-induced ER stress, inflammation, and apoptosis in renal cells, in part, by regulating the PI3K/AKT and PERK-eIF2α-ATF4 signaling pathways.
Collapse
Affiliation(s)
- Xiao-meng Wei
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
| | - Shuang Jiang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Shan-shan Li
- Institute
of Special Wild Economic Animals and Plant, Chinese Academy of Agricultural Sciences, Changchun 132109, China
| | - Yin-shi Sun
- Institute
of Special Wild Economic Animals and Plant, Chinese Academy of Agricultural Sciences, Changchun 132109, China
| | - Shi-han Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Wen-cong Liu
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Zi Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Ying-ping Wang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Rui Zhang
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| | - Wei Li
- College
of Chinese Medicinal Materials, Jilin Agricultural
University, Changchun 130118, China
- National
& Local Joint Engineering Research Center for Ginseng Breeding
and Development, Changchun 130118, China
| |
Collapse
|
34
|
He XL, Xu XH, Shi JJ, Huang M, Wang Y, Chen X, Lu JJ. Anticancer Effects of Ginsenoside Rh2: A Systematic Review. Curr Mol Pharmacol 2021; 15:179-189. [PMID: 33687905 DOI: 10.2174/1874467214666210309115105] [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: 09/28/2020] [Revised: 12/22/2020] [Accepted: 01/18/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND As one of the effective pharmacological constituents of Ginseng Radix et Rhizoma, ginsenoside Rh2 (Rh2) exerts a remarkable anticancer effect on various cancer cell lines in vitro and strongly inhibits tumor growth in vivo without severe toxicity. OBJECTIVE This article reviewed existing evidence supporting the anticancer effects of Rh2 to classify and conclude previous and current knowledge on the mechanisms and therapeutic effects of Rh2, as well as to promote the clinical application of this natural product. CONCLUSION This article reviewed the anticancer efficacies and mechanisms of Rh2, including the induction of cell cycle arrest and programmed cell death, repression of metastasis, alleviation of drug resistance, and regulation of the immune system. Finally, this paper discussed the research and application prospects of Rh2.
Collapse
Affiliation(s)
- Xin-Ling He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Xiao-Huang Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Jia-Jie Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Mingqing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122. China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao. China
| |
Collapse
|
35
|
Wen T, Song L, Hua S. Perspectives and controversies regarding the use of natural products for the treatment of lung cancer. Cancer Med 2021; 10:2396-2422. [PMID: 33650320 PMCID: PMC7982634 DOI: 10.1002/cam4.3660] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer‐related mortality both in men and women and accounts for 18.4% of all cancer‐related deaths. Although advanced therapy methods have been developed, the prognosis of lung cancer patients remains extremely poor. Over the past few decades, clinicians and researchers have found that chemical compounds extracted from natural products may be useful for treating lung cancer. Drug formulations derived from natural compounds, such as paclitaxel, doxorubicin, and camptothecin, have been successfully used as chemotherapeutics for lung cancer. In recent years, hundreds of new natural compounds that can be used to treat lung cancer have been found through basic and sub‐clinical research. However, there has not been a corresponding increase in the number of drugs that have been used in a clinical setting. The probable reasons may include low solubility, limited absorption, unfavorable metabolism, and severe side effects. In this review, we present a summary of the natural compounds that have been proven to be effective for the treatment of lung cancer, as well as an understanding of the mechanisms underlying their pharmacological effects. We have also highlighted current controversies and have attempted to provide solutions for the clinical translation of these compounds.
Collapse
Affiliation(s)
- Tingting Wen
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Lei Song
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Shucheng Hua
- Department of Respiratory Medicine, Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|
36
|
Hong H, Baatar D, Hwang SG. Anticancer Activities of Ginsenosides, the Main Active Components of Ginseng. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8858006. [PMID: 33623532 PMCID: PMC7875636 DOI: 10.1155/2021/8858006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022]
Abstract
Cancer incidence rate has been increasing drastically in recent years. One of the many cancer treatment methods is chemotherapy. Traditional medicine, in the form of complementary and alternative therapy, is actively used to treat cancer, and many herbs and active ingredients of such therapies are being intensely studied to integrate them into modern medicine. Ginseng is traditionally used as a nourishing tonic and for treating various diseases in Asian countries. The therapeutic potential of ginseng in modern medicine has been studied extensively; the main bioactive component of ginseng is ginsenosides, which have gathered attention, particularly for their prospects in the treatment of fatal diseases such as cancer. Ginsenosides displayed their anticancer and antimetastatic properties not only via restricting cancer cell proliferation, viability, invasion, and migration but also by promoting apoptosis, cell cycle arrest, and autophagy in several cancers, such as breast, brain, liver, gastric, and lung cancer. Additionally, ginsenosides can work synergistically with already existing cancer therapies. Thus, ginsenosides may be used alone or in combination with other pharmaceutical agents in new therapeutic strategies for cancer. To date however, there is little systematic summary available for the anticancer effects and therapeutic potential of ginsenosides. Therefore, we have reviewed and discussed all available literature in order to facilitate further research of ginsenosides in this manuscript.
Collapse
Affiliation(s)
- Heeok Hong
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Delgerzul Baatar
- Laboratory of Genetics, Institute of Biology, Mongolian Academy of Sciences, Peace Avenue 13330, Ulaanbaatar, Mongolia
| | - Seong Gu Hwang
- Department of Animal Life and Environmental Science, Hankyong National University, Anseong City 17579, Republic of Korea
| |
Collapse
|
37
|
Chen Y, Zhang Y, Song W, Zhang Y, Dong X, Tan M. Ginsenoside Rh2 Improves the Cisplatin Anti-tumor Effect in Lung Adenocarcinoma A549 Cells via Superoxide and PD-L1. Anticancer Agents Med Chem 2021; 20:495-503. [PMID: 31814556 DOI: 10.2174/1871520619666191209091230] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/21/2019] [Accepted: 10/31/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ginsenoside Rh2 (Rh2) is a major biological component of ginseng that exerts antitumor activities in multiple cancers including Non-Small Cell Lung Cancers (NSCLCs). Rh2 also enhances the anti-tumor effects of various chemotherapy drugs including cisplatin at relatively low concentrations. Here, the mechanistic role of Rh2 in chemotherapy-treated NSCLCs will be investigated. METHODS In this study, FACS, western blot and siRNA addition were used to analyze the role of Rh2 in cisplatin- treated lung adenocarcinoma A549 and H1299 cells. RESULTS Subsequent observations indicated that Rh2 enhanced cisplatin-induced NSCLCs A549 and H1299 cells apoptosis. Cisplatin-induced productive autophagy was repressed by Rh2 in A549 cells. Rh2 also enhanced cisplatin cytotoxicity by elevating superoxide dismutase activity and repressing cisplatin-induced superoxide generation. Conversely, Rh2 was found to repress cisplatin-induced phosphorylation of epidermal growth factor receptor, phosphoinositide 3-kinase, protein kinase B, and autophagy. Cisplatin-induced Programmed Death- Ligand 1 (PD-L1) expression was repressed by Rh2 via the superoxide. CONCLUSION These findings suggest that Rh2 enhanced the function of cisplatin by repressing superoxide generation, PD-L1 expression, and autophagy in lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110042, Liaoning Province, China
| | - Yuqiang Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110042, Liaoning Province, China
| | - Wei Song
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110042, Liaoning Province, China
| | - Ying Zhang
- Oncology Medicine Department, Shengjing Hospital of China Medical University, Shenyang, 110042, Liaoning Province, China
| | - Xiu Dong
- School of Preclinical Medicine, Liaoning University of Traditional Chinese Medicine, 79 Chong Shan Dong Lu, Huanggu District, Shenyang, Liaoning, 110847, China
| | - Mingqi Tan
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110042, Liaoning Province, China
| |
Collapse
|
38
|
Ni M, Wang H, Wang M, Zhou W, Zhang J, Wu J, Zhang D, Jing Z, Liu X, Wu Z, Guo S, Jia S, Zhang X, Sheng X. Investigation on the Efficiency of Chinese Herbal Injections for Treating Non-small Cell Lung Cancer With Vinorelbine and Cisplatin Based on Multidimensional Bayesian Network Meta-Analysis. Front Pharmacol 2021; 11:631170. [PMID: 33708126 PMCID: PMC7941272 DOI: 10.3389/fphar.2020.631170] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Background: As non-small cell lung cancer (NSCLC) seriously threatens human health, several clinical studies have reported that Chinese herbal injections (CHIs) combined with vinorelbine and cisplatin (NP) are beneficial. This multidimensional network meta-analysis was performed to explore the preferable options among different CHIs for treating NSCLC. Methods: A literature search was performed in several databases to identify randomized controlled trials (RCTs) of CHIs in the treatment of NSCLC from inception to January 31, 2019. Final included studies met the eligibility criteria and methodological quality recommendations. Data analysis was performed using Stata 13.0 and WinBUGS 14.0 software. Each outcome was presented as an odds ratio and the surface under the cumulative ranking curve value (SCURA). The “scatterplot3d” package in R 3.6.1 software was used to perform multidimensional cluster analysis. Results: Ultimately, 97 eligible RCTs involving 7,440 patients and 14 CHIs were included in this network meta-analysis. Combined with NP chemotherapy, Kanglaite injection plus NP exhibited a better impact on the clinical effectiveness rate (SCURA probability: 78.34%), and Javanica oil emulsion injection plus NP was better in the performance status (95.44%). Huachansu injection plus NP was dominant in reducing thrombocytopenia (92.67%) and gastrointestinal reactions (92.52%). As to multidimensional cluster analysis, Shenmai injection plus NP was superior considering improving the clinical effectiveness rate, performance status and relieving leukopenia. Conclusions: The combination of CHIs and NP has a better impact on patients with NSCLC than NP alone. Among them, Shenmai injection plus NP, Kanglaite injection plus NP and Javanica oil emulsion injection plus NP were notable. Nevertheless, more multicenter and better designed RCTs are needed to validate our findings.
Collapse
Affiliation(s)
- Mengwei Ni
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haojia Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Miaomiao Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiwei Jing
- China Academy of Chinese Medicine Sciences, Beijing, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Siyu Guo
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shanshan Jia
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomeng Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoguang Sheng
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
39
|
Yang Z, Huang Y, Zhu L, Yang K, Liang K, Tan J, Yu B. SIRT6 promotes angiogenesis and hemorrhage of carotid plaque via regulating HIF-1α and reactive oxygen species. Cell Death Dis 2021; 12:77. [PMID: 33436551 PMCID: PMC7804142 DOI: 10.1038/s41419-020-03372-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
As a member of Sirtuins family, SIRT6 participates in the physiological and pathological progress of DNA repair, anti-aging, metabolism, and so on. Several studies have demonstrated that knockdown of SIRT6 inhibited the development of atherosclerosis (AS), indicated SIRT6 as a protective factor for AS. However, we confirmed SIRT6 was significantly overexpressed in human unstable carotid plaques compared with stable carotid plaques. This result indicated a more complex role of SIRT6 in AS. Furthermore, we constructed mice model with unstable carotid plaque and injected them with SIRT6 overexpressed adeno-associated virus (AAV-SIRT6). AAV-SIRT6 significantly promoted angiogenesis as well as hemorrhage in plaques. In vitro, we demonstrated overexpression of SIRT6 prevented HIF-1α from degradation by deubiquitination at K37 and K532 of HIF-1α, thus promoted the expression of HIF-1α under both normoxia and hypoxia in human umbilical vein endothelial cells (HUVECs). Through regulating HIF-1α, overexpression of SIRT6 promoted invasion, migration, proliferation, as well as tube formation ability of HUVECs. Interestingly, under different conditions, SIRT6 played different roles in the function of HUVECs. Under oxidative stress, another important pathological environment for AS, SIRT6 bound to the promoter of Catalase, a main reactive oxygen species scavenger, and depleted H3K56 acetylation, thus inhibited expression and activity of Catalase at the transcriptional level. Subsequently, inhibited Catalase promoted reactive oxygen species (ROS) under oxidative stress. Accumulated ROS further aggravated oxidative stress injury of HUVECs. On one hand, SIRT6 promoted angiogenesis in plaque via HIF-1α under hypoxia. On the other hand, SIRT6 promoted injury of neovascular via ROS under oxidative stress. It is this process of continuous growth and damage that leads to hemorrhage in carotid plaque. In conclusion, we innovatively confirmed SIRT6 promoted the angiogenesis and IPH via promoting HIF-1α and ROS in different environments, thus disclosed the unknowing danger of SIRT6.
Collapse
Affiliation(s)
- Zhou Yang
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Yijun Huang
- Department of General Surgery, Huashan Hospital North, Fudan University, Shanghai, 201907, China
| | - Lei Zhu
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Kai Yang
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Kun Liang
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jinyun Tan
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Bo Yu
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
40
|
Wan X, Li X, Liu D, Gao X, Chen Y, Chen Z, Fu C, Lin L, Liu B, Zhao C. Physicochemical characterization and antioxidant effects of green microalga Chlorella pyrenoidosa polysaccharide by regulation of microRNAs and gut microbiota in Caenorhabditis elegans. Int J Biol Macromol 2020; 168:152-162. [PMID: 33301848 DOI: 10.1016/j.ijbiomac.2020.12.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022]
Abstract
A novel polysaccharide from Chlorella pyrenoidosa (CPP) was separated and purified with the average molecular weight 15.8 kDa. It was composed of seven monosaccharides including mannose, rhamnose, glucuronic acid, galacturonic acid, glucose, galactose, and arabinose. FT-IR and NMR spectra analysis further revealed that CPP was an acidic polysaccharide consisting of β-L-Arap-(1→, →2)-α-L-Rhap-(1→, β-D-GlcpA-(1→, →4)-α-D-GalpA-(1→, →6)-β-D-Glcp-(1→, →3)-β-D-Manp-(1→, and →3, 6)-β-D-Galp-(1→. The CPP treatment could effectively prolong lifespan of Caenorhabditis elegans under the oxidative stress conditions and inhibit the accumulation of reactive oxygen species (ROS) and malondialdehyde (MDA) as well as enhancing the level of superoxide dismutase (SOD). It could up-regulate the expressions of Daf-16 and Skn-1 genes via declining miR-48-3p, miR-48-5p, and miR-51-5p translocation. Moreover, 16S rRNA sequencing revealed that the CPP-enriched Faecalibacterium, Haemophilus, Vibrio, and Shewanella were strongly correlated with SOD, MDA, apoptosis, and ROS. These results indicated that CPP may be considered as a desired ingredient on regulating the aging and oxidative diseases.
Collapse
Affiliation(s)
- Xuzhi Wan
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoqing Li
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Dan Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoxiang Gao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yihan Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zhengxin Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Caili Fu
- Beijing Key Laboratory of Flavor Chemistry, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Luan Lin
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou 362000, China
| | - Bin Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Engineering Research Center of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China
| | - Chao Zhao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Engineering Research Center of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
41
|
Jin Y, Huynh DTN, Nguyen TLL, Jeon H, Heo KS. Therapeutic effects of ginsenosides on breast cancer growth and metastasis. Arch Pharm Res 2020; 43:773-787. [PMID: 32839835 DOI: 10.1007/s12272-020-01265-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/13/2020] [Indexed: 12/16/2022]
Abstract
Breast cancer is the most common cause of cancer-related deaths among women worldwide. Thus, the development of new and effective low-toxicity drugs is vital. The specific characteristics of breast cancer have allowed for the development of targeted therapy towards each breast cancer subtype. Nevertheless, increasing drug resistance is displayed by the changing phenotype and microenvironments of the tumor through mutation or dysregulation of various mechanisms. Recently, emerging data on the therapeutic potential of biocompounds isolated from ginseng have been reported. Therefore, in this review, various roles of ginsenosides in the treatment of breast cancer, including apoptosis, autophagy, metastasis, epithelial-mesenchymal transition, epigenetic changes, combination therapy, and drug delivery system, have been discussed.
Collapse
Affiliation(s)
- Yujin Jin
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Diem Thi Ngoc Huynh
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Thuy Le Lam Nguyen
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Hyesu Jeon
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea. .,Institute of Drug Research & Development, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
42
|
Li X, Chu S, Lin M, Gao Y, Liu Y, Yang S, Zhou X, Zhang Y, Hu Y, Wang H, Chen N. Anticancer property of ginsenoside Rh2 from ginseng. Eur J Med Chem 2020; 203:112627. [PMID: 32702586 DOI: 10.1016/j.ejmech.2020.112627] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
Ginseng has been used as a well-known traditional Chinese medicine since ancient times. Ginsenosides as its main active constituents possess a broad scope of pharmacological properties including stimulating immune function, enhancing cardiovascular health, increasing resistance to stress, improving memory and learning, developing social functioning and mental health in normal persons, and chemotherapy. Ginsenoside Rh2 (Rh2) is one of the major bioactive ginsenosides from Panax ginseng. When applied to cancer treatment, Rh2 not only exhibits the anti-proliferation, anti-invasion, anti-metastasis, induction of cell cycle arrest, promotion of differentiation, and reversal of multi-drug resistance activities against multiple tumor cells, but also alleviates the side effects after chemotherapy or radiotherapy. In the past decades, nearly 200 studies on Rh2 in the treatment of cancer have been published, however no specific reviews have been conducted by now. So the purpose of this review is to provide a systematic summary and analysis of the anticancer effects and the potential mechanisms of Rh2 extracted from Ginseng then give a future prospects about it. In the end of this paper the metabolism and derivatives of Rh2 also have been documented.
Collapse
Affiliation(s)
- Xun Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China; Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, PR China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Shifeng Chu
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Meiyu Lin
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Yan Gao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yingjiao Liu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Xin Zhou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yani Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Yaomei Hu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Huiqin Wang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Naihong Chen
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, PR China; Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, PR China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China; Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China.
| |
Collapse
|
43
|
Han Q, Han L, Tie F, Wang Z, Ma C, Li J, Wang H, Li G. (20S)-Protopanaxadiol Ginsenosides Induced Cytotoxicity via Blockade of Autophagic Flux in HGC-27 Cells. Chem Biodivers 2020; 17:e2000187. [PMID: 32384197 DOI: 10.1002/cbdv.202000187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022]
Abstract
(20S)-Protopanaxadiol ginsenosides Rg3, Rh2 and PPD have been demonstrated for their anticancer activity. However, the underlying mechanism of their antitumor activity remains unclear. In the present study, we investigated the role of these three ginsenosides on cell proliferation and death of human gastric cancer cells (HGC-27 cells). The sulforhodamine B (SRB) assay, Western blot analysis, fluorescence microscopy, confocal microscopy, high performance liquid chromatography (HPLC) analysis, flow cytometry, and transmission electron microscopy (TEM) were used to evaluate cell proliferation, apoptosis, and autophagy. The results showed that both Rh2 and PPD were more effective than Rg3 in inhibiting HGC-27 cell proliferation and inducing cytoplasmic vacuolation, while no significant changes in apoptosis were observed. Interestingly, cytoplasmic vacuolation and blockade of autophagy flux were observed after treatment with Rh2 and PPD. Rh2 obviously up-regulated the expression of the LC3II and p62. Furthermore, the increase in lysosomal pH and membrane rupture was observed in Rh2-treated and PPD-treated cells. When HGC-27 cells were pretreated with bafilomycin A1, a specific inhibitor of endosomal acidification, cellular vacuolization was increased, and the cell viability was significantly decreased, which indicated that Rh2-induced lysosome-damage accelerated cell death. Furthermore, data derived from mitochondrial analysis showed that excessive mitochondrial reactive oxygen species (ROS) and dysregulation of mitochondrial energy metabolism were caused by Rh2 and PPD treatment in HGC-27 cells. Taken together, these phenomena indicated that Rh2 and PPD inhibited HCG-27 cells proliferation by inducing mitochondria damage, dysfunction of lysosomes, and blockade of autophagy flux. The number of glycosyl groups at C-3 position could have an important effect on the cytotoxicity of Rg3, Rh2 and PPD.
Collapse
Affiliation(s)
- Qingqing Han
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| | - Lijuan Han
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, 810016, P. R. China
| | - Fangfang Tie
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, P. R. China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| | - Chengjun Ma
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| | - Ji Li
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| | - Honglun Wang
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China.,Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, P. R. China
| | - Gang Li
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| |
Collapse
|
44
|
Lu H, Yuan X, Zhang Y, Han M, Liu S, Han K, Liang P, Cheng J. HCBP6 deficiency exacerbates glucose and lipid metabolism disorders in non-alcoholic fatty liver mice. Biomed Pharmacother 2020; 129:110347. [PMID: 32535386 DOI: 10.1016/j.biopha.2020.110347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/23/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), which often accompanied by metabolic syndrome, such as obesity, diabetes and dyslipidemia, has become a global health problem. Our previous results show that HCV core protein binding protein 6 (HCBP6) could maintain the triglyceride homeostasis in liver cells. However, the role of HCBP6 in NAFLD and its associated metabolic disorders remains incompletely understood. METHODS Hepatic HCBP6 expression was determined by qRT-PCR, Western blot and immunohistochemistry analysis. HCBP6 knockout (HCBP6-KO) mice were constructed and fed a high-fat diet (HFD) to induce NAFLD. The effects of HCBP6 on glucose and lipid metabolism were measured by HE staining, qRT-PCR, Western blot and GTT. Wild-type and HCBP6-KO mice kept on a HFD were treated with ginsenosides Rh2, and HE staining and GTT were used to study the function of Rh2 in metabolism disorders. RESULTS HCBP6 is reduced in HFD-fed mice. HCBP6 deficiency increased the body weight, aggravated fatty liver and deteriorated lipid homeostasis as well as glucose homeostasis in HFD-induced mouse model of NAFLD. Moreover, HCBP6-KO mice failed to maintain body temperature upon cold challenge. Mechanistically, HCBP6 could regulate lipolysis and fatty acid oxidation via activation of AMKP in vivo. In addition, HCBP6 expression was upregulated by ginsenosides Rh2. Accordingly, ginsenosides Rh2 administrations improved HFD-induced fatty liver and glucose tolerance. CONCLUSIONS These findings indicated that HCBP6 is essential in maintaining lipid and glucose homeostasis and body temperature. HCBP6 augmented by ginsenosides Rh2 may be a promising therapeutic strategy for the treatment of metabolic disorders in NAFLD mice.
Collapse
Affiliation(s)
- Hongping Lu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Xiaoxue Yuan
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China.
| | - Yu Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Department of Hepatology Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ming Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Shunai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Kai Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Pu Liang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China
| | - Jun Cheng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; Beijing Key Laboratory of Emerging Infectious Diseases, Beijing 100015, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing 100191, China.
| |
Collapse
|
45
|
Hu X, Zhang ZY, Wu LW, Zeng LH, Chen H, Zhu HJ, Zhang JK, Shao J, Zhang C, Li YL, Lin NM. A natural anthraquinone derivative shikonin synergizes with AZD9291 against wtEGFR NSCLC cells through reactive oxygen species-mediated endoplasmic reticulum stress. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 68:153189. [PMID: 32070867 DOI: 10.1016/j.phymed.2020.153189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/18/2020] [Accepted: 02/07/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND NSCLC is the major type of lung cancer and the survival rates of NSCLC patients remain low. AZD9291 is a third-generation EGFR-TKI and approved to treat NSCLC patients harboring EGFR T790M mutation and common targetable activating EGFR mutations, but it has a limited effect for wtEGFR NSCLC. PURPOSE The current study investigated whether shikonin could enhance the antitumor effect of AZD9291 in wtEGFR NSCLC cells. METHODS SRB and colony formation assay were used to detect the proliferation of NSCLC cells, propidium iodide staining was performed to detect the apoptosis, ROS was analyzed using DCFH-DA staining, and western blot was used to detect the expression of indicated proteins. RESULTS We demonstrated that shikonin, a natural ROS inducer, could enhance the antitumor effect of AZD9291 in wtEGFR NSCLC cells. In addition, shikonin increased AZD9291-induced apoptosis accompanying with the generation of ROS and activation of ER stress. Furthermore, ROS inhibition by NAC or GSH reversed the apoptosis induced by shikonin plus AZD9291, and recovered the ER stress activated by combination treatment, indicating that ROS mediated ER stress played a vital role in this combination therapy. Moreover, shikonin increased the anticancer activity of AZD9291 in primary wtEGFR NSCLC cells through ROS-mediated ER stress. CONCLUSION Our study suggests that combining shikonin with AZD9291 is a promising therapeutic strategy for treating wtEGFR NSCLC patients.
Collapse
Affiliation(s)
- Xiu Hu
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China,; College of Pharmaceutical Sciences, Zhejiang University, No.866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Zuo-Yan Zhang
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China,; College of Pharmaceutical Sciences, Zhejiang University, No.866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Lin-Wen Wu
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China,; College of Pharmaceutical Sciences, Zhejiang University, No.866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, China
| | - Ling-Hui Zeng
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China
| | - Hui Chen
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China
| | - Hua-Jian Zhu
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China
| | - Jian-Kang Zhang
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China
| | - Jiaan Shao
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China
| | - Chong Zhang
- School of Medicine, Zhejiang University City College, No.51 Huzhou Street, Hangzhou, Zhejiang, 310015, China,.
| | - Yang-Ling Li
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No.261 Huansha Road, Hangzhou, Zhejiang, 310006, China.
| | - Neng-Ming Lin
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, No.261 Huansha Road, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
46
|
Yang L, Zou H, Gao Y, Luo J, Xie X, Meng W, Zhou H, Tan Z. Insights into gastrointestinal microbiota-generated ginsenoside metabolites and their bioactivities. Drug Metab Rev 2020; 52:125-138. [PMID: 31984805 DOI: 10.1080/03602532.2020.1714645] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gastrointestinal microbiota and host co-evolve into a complex 'super-organism,' and this relationship plays a vital role in many physiological processes, such as drug metabolism. Ginseng is an important medicinal resource and the main ingredients are ginsenosides, which are less polar, difficult to absorb, and have low bioavailability. However, studies have shown that the biological activity of ginsenosides such as compound K (CK), ginsenoside Rg3 (Rg3), ginsenoside Rh2 (Rh2), 20(S)-protopanaxatriol (20(S)-PPT), and 20(S)-protopanaxadiol (20(S)-PPD) is closely related to the gastrointestinal microbiota. In this paper, the metabolic pathway of gastrointestinal microbiota-generated ginsenosides and the main pharmacological effects of these metabolites are discussed. Furthermore, our study provides a new insight into the discovery of novel drugs. Specifically, in new drug screening process, candidates with low biological activity and bioavailability should not be excluded. Because their metabolites may exhibit good pharmacological effects due to the involvement of the gastrointestinal microbiota. In addition, in further research studies to develop probiotics, a combination of agents could exert greater efficacy than single agents. Moreover, differences in lifestyle and diet lead to differences in the gastrointestinal microbiota in the human body. Therefore, administration of the same drug dose to different individuals could elicit different therapeutic effects, owing to the involvement of the gastrointestinal microbiota. Thus, treatment accuracy could be achieved by detecting the gastrointestinal microbiota before drug treatment.HighlightsGastrointestinal microbiota plays a decisive role in bioactivities of ginsenosides.The metabolic pathway and main pharmacological effects of ginsenoside metabolites are discussed.It provides new insights into novel drug discovery and further research to find probiotic, combinations to exert greater efficacy.Differences in lifestyle and diet, varies the gastrointestinal microbiota in the human body. However, the same dose of a drug producing different therapeutic effects may involve gastrointestinal microbiota.
Collapse
Affiliation(s)
- Li Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Hecun Zou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China.,Institute of Life Sciences, Chongqing Medical University, Chongqing, Hunan, PR China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Junjia Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Xiaonv Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Wenhui Meng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, PR China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, PR China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, PR China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, PR China
| |
Collapse
|
47
|
Yun M, Yi YS. Regulatory roles of ginseng on inflammatory caspases, executioners of inflammasome activation. J Ginseng Res 2019; 44:373-385. [PMID: 32372859 PMCID: PMC7195600 DOI: 10.1016/j.jgr.2019.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/06/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an immune response that protects against pathogens and cellular stress. The hallmark of inflammatory responses is inflammasome activation in response to various stimuli. This subsequently activates downstream effectors, that is, inflammatory caspases such as caspase-1, 4, 5, 11, and 12. Extensive efforts have been made on developing effective and safe anti-inflammatory therapeutics, and ginseng has long been traditionally used as efficacious and safe herbal medicine in treating various inflammatory and inflammation-mediated diseases. Many studies have successfully shown that ginseng plays an anti-inflammatory role by inhibiting inflammasomes and inflammasome-activated inflammatory caspases. This review discusses the regulatory roles of ginseng on inflammatory caspases in inflammatory responses and also suggests new research areas on the anti-inflammatory function of ginseng, which provides a novel insight into the development of ginseng as an effective and safe anti-inflammatory herbal medicine.
Collapse
Key Words
- AIM2, Absent in melanoma 2
- ASC, Apoptosis-associated speck-like protein containing CARD
- CARD, C-terminal caspase recruit domain
- COX-2, Cyclooxygenase-2
- Caspase, Cysteine aspartate–specific protease
- DAMP, Danger-associated molecular pattern
- FIIND, Functional-to-find domain
- GSDMD, Gasdermin D
- Ginseng
- Ginsenoside
- HIN, Hematopoietic interferon-inducible nuclear protein
- IL, Interleukin
- Inflammasome
- Inflammation
- Inflammatory caspase
- LPS, Lipopolysaccharide
- LRR, Leucine-rich repeat
- NACHT, Nucleotide-binding and oligomerization domain
- NF-κB, Nuclear factor-kappa B
- NLR, Nucleotide-binding oligomerization domain-like receptor
- NO, Nitric oxide
- PAMP, Pathogen-associated molecular pattern
- PGE2, Prostaglandin E2
- PRR, Pattern-recognition receptor
- PYD, N-terminal pyrin domain
- RGE, Korean Red Ginseng
- ROS, Reactive oxygen species
Collapse
Affiliation(s)
- Miyong Yun
- Department of Bioindustry and Bioresource Engineering, Sejong University, Seoul, Republic of Korea
| | - Young-Su Yi
- Department of Life Science, Kyonggi University, Suwon, Republic of Korea
| |
Collapse
|
48
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 315] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
49
|
Wang J, Zhang Y, Liu X, Wang J, Li B, Liu Y, Wang J. Alantolactone enhances gemcitabine sensitivity of lung cancer cells through the reactive oxygen species-mediated endoplasmic reticulum stress and Akt/GSK3β pathway. Int J Mol Med 2019; 44:1026-1038. [PMID: 31524219 PMCID: PMC6657978 DOI: 10.3892/ijmm.2019.4268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer‑associated mortality in China and globally. Gemcitabine (GEM), as a first‑line therapeutic drug, has been used to treat lung cancer, but GEM resistance poses a major limitation on the efficacy of GEM chemotherapy. Alantolactone (ALT), a sesquiterpene lactone compound isolated from Inula helenium, has been identified to exert anticancer activity in various types of cancer, including breast, pancreatic, lung squamous and colorectal cancer. However, the underlying mechanisms of the anticancer activity of ALT in lung cancer remain to be fully elucidated. The present study aimed to determine whether ALT enhances the anticancer efficacy of GEM in lung cancer cells and investigated the underlying mechanisms. The cell viability was assessed with a Cell Counting Kit‑8 assay. The cell cycle, apoptosis and the level of reactive oxygen species (ROS) were assessed by flow cytometry, and the expression of cell cycle‑associated and apoptosis‑associated proteins were determined by western blot analysis. The results demonstrated that ALT inhibited cell growth and induced S‑phase arrest and cell apoptosis in A549 and NCI‑H520 cells. Furthermore, ALT increased the level of ROS, inhibited the Akt/glycogen synthase kinase (GSK)3β pathway and induced endoplasmic reticulum (ER) stress in A549 and NCI‑H520 cells. Additionally, ALT treatment sensitized lung cancer cells to GEM. Analysis of the molecular mechanisms further revealed that ALT enhanced the anticancer effects of GEM via ROS‑mediated activation of the Akt/GSK3β and ER stress pathways. In conclusion, combined treatment with ALT and GEM may have potential as a clinical strategy for lung cancer treatment.
Collapse
Affiliation(s)
| | | | - Xu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Jizhao Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| | - Bin Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yongkang Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Jiansheng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061
| |
Collapse
|
50
|
Zheng SW, Xiao SY, Wang J, Hou W, Wang YP. Inhibitory Effects of Ginsenoside Ro on the Growth of B16F10 Melanoma via Its Metabolites. Molecules 2019; 24:E2985. [PMID: 31426477 PMCID: PMC6721120 DOI: 10.3390/molecules24162985] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022] Open
Abstract
Ginsenoside Ro (Ro), a major saponin derived and isolated from Panax ginseng C.A. Meyer, exerts multiple biological activities. However, the anti-tumour efficacy of Ro remains unclear because of its poor in vitro effects. In this study, we confirmed that Ro has no anti-tumour activity in vitro. We explored the anti-tumour activity of Ro in vivo in B16F10 tumour-bearing mice. The results revealed that Ro considerably suppressed tumour growth with no significant side effects on immune organs and body weight. Zingibroside R1, chikusetsusaponin IVa, and calenduloside E, three metabolites of Ro, were detected in the plasma of Ro-treated tumour-bearing mice and showed excellent anti-tumour effects as well as anti-angiogenic activity. The results suggest that the metabolites play important roles in the anti-tumour efficacy of Ro in vivo. Additionally, the haemolysis test demonstrated that Ro has good biocompatibility. Taken together, the findings of this study demonstrate that Ro markedly suppresses the tumour growth of B16F10-transplanted tumours in vivo, and its anti-tumour effects are based on the biological activity of its metabolites. The anti-tumour efficacy of these metabolites is due, at least in part, to its anti-angiogenic activity.
Collapse
Affiliation(s)
- Si-Wen Zheng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Sheng-Yuan Xiao
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China
| | - Jia Wang
- School of Pharmaceutical Sciences Changchun University of Chinese Medicine, Changchun 130117, China
| | - Wei Hou
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China
| | - Ying-Ping Wang
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China.
- National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, China.
| |
Collapse
|