1
|
Niu X, Gu H, Li J, Zuo J, Ren W, Huang Y, Shu X, Jiang C, Shu P. Efficacy and safety of Atractylodes macrocephala-containing traditional Chinese medicine combined with neoadjuvant chemotherapy in the treatment of advanced gastric cancer: a systematic evaluation and meta-analysis. Front Oncol 2024; 14:1431381. [PMID: 39479020 PMCID: PMC11521787 DOI: 10.3389/fonc.2024.1431381] [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: 05/11/2024] [Accepted: 09/18/2024] [Indexed: 11/02/2024] Open
Abstract
Background In China, Atractylodes-containing Chinese medicines are widely used as adjuvant therapy to neoadjuvant chemotherapy (NAC) in individuals diagnosed with advanced gastric cancer (AGC). Nevertheless, the findings concerning its effectiveness are still restricted. The aim from this research was to examine the efficiency and security Atractylodes macrocephala-containing traditional Chinese medicine together with NAC in the management of AGC. Methods Literature was systematically searched across 8 electronic databases until September 20, 2023. Two researchers conducted a thorough review of the selected studies. The primary outcome measures included the objective response rate (ORR), disease control rate (DCR), quality of life (QOL), adverse drug reactions (ADRs), and levels of peripheral blood lymphocytes. The relevant effect estimates are as follows as risk ratios (RR) or mean differences (MD) with corresponding 95% confidence intervals (CI). Credibility of information was evaluated using the GRADE analyzer. Results The results showed that solely on the basis of the accessible literature examined in NAC patients, individuals who received the therapeutic regimen containing Atractylodis Macrocephalae Chinese herbal preparations demonstrated a superior overall response rate (Relative Risk: 1.41, 95% confidence interval: 1.27-1.57, P < 0.001); DCR (RR: 1.20, 95% confidence interval: 1.13-1.27, P < 0.001), as compared to QOL (RR: 1.43, 95% confidence interval: 1.30-1.57, P < 0.001, MD: 8.47, 95% confidence interval: 7.16 - 9.77, P < 0.001); the proportions of CD3+ T-cells, CD4+ T-cells, CD8+ T-cells, CD4+CD8+ T-cells were increased; and the incidence of adverse reactions was decreased. Subgroup analyses showed that oral administration of all the traditional Chinese medicines containing Atractylodes macrocephala could improve tumor efficacy. Regardless of the duration of therapy of ≥8 weeks or <8 weeks, Atractylodes macrocephala-containing traditional Chinese medicine increased the tumor response in AGC patients. Combination of Atractylodes macrocephala-containing TCM with neoadjuvant chemotherapy increased ORR and DCR; when used in conjunction with cisplatin, only ORR was increased. Conclusion The combination of Atractylodes macrocephala-containing herbs with NAC in the treatment of AGC improves efficacy, improves prognosis, and reduces adverse effects. Nevertheless, additional high-quality randomized trials are required. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023461079.
Collapse
Affiliation(s)
- Xiaotao Niu
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Haoqing Gu
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jingzhan Li
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiaqian Zuo
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenqin Ren
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yujie Huang
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xinyan Shu
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chao Jiang
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Peng Shu
- Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, China
- School of No. 1 Clinical Medical, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Lin Y, Chen K, Zhu M, Song W, Wu G, Pan A. Atractylenolide II regulates the proliferation, ferroptosis, and immune escape of hepatocellular carcinoma cells by inactivating the TRAF6/NF-κB pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7697-7710. [PMID: 38709266 DOI: 10.1007/s00210-024-03046-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/09/2024] [Indexed: 05/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common and lethal tumor worldwide. Atractylenolide II (AT-II) is a natural sesquiterpenoid monomer, with anti-tumor effect. To address the effect and mechanisms of AT-II on HCC. The role and mechanisms of AT-II were assessed through cell counting kit-8, flow cytometry, enzyme-linked immunosorbent assay, immunofluorescence, and western blot experiments in Hep3B and Huh7 cells. In vivo experiments were conducted in BALB/c nude mice using immunohistochemistry and western blot assays. AT-II decreased the cell viability of Hep3B and Huh7 cells with a IC50 of 96.43 µM and 118.38 µM, respectively. AT-II increased relative Fe2+ level, which was further promoted with the incubation of erastin and declined with the ferrostatin-1 in Hep3B and Huh7 cells. AT-II enhanced the level of ROS and MDA, but reduced the GSH level, and the expression of xCT and GPX4. AT-II elevated the percent of CD8+ T cells and the IFN-γ contents, and declined the IL-10 concentrations and the expression of PD-L1 in Hep3B and Huh7 cells. AT-II downregulated the relative protein level of TRAF6, p-p65/p-65, and p-IkBα/IkBα, which was rescued with overexpression of TRAF6. Upregulation of TRAF6 also reversed the effect of AT-II on proliferation, ferroptosis, and immune escape in Hep3B cells. In vivo, AT-II reduced tumor volume and weight, the level of GPX4, xCT, and PD-L1, and the expression of TRAF6, p-p65/p-65, and p-IkBα/IkBα, with the increased expression of CD8. AT-II modulated the proliferation, ferroptosis, and immune escape of HCC cells by downregulating the TRAF6/NF-κB pathway.
Collapse
Affiliation(s)
- Yujie Lin
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University, Yuexiu District, No. 107, Yanjiang West Road, Guangzhou Guangdong Province, 510120, China
| | - Ke Chen
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University, Yuexiu District, No. 107, Yanjiang West Road, Guangzhou Guangdong Province, 510120, China
| | - Min Zhu
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University, Yuexiu District, No. 107, Yanjiang West Road, Guangzhou Guangdong Province, 510120, China
| | - Wei Song
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University Shenshan Central Hospital, Shanwei, 516600, China
| | - Guiyun Wu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University, Yuexiu District, No. 107, Yanjiang West Road, Guangzhou Guangdong Province, 510120, China.
| | - Aizhen Pan
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital Affiliated to Sun Yat-sen University, Yuexiu District, No. 107, Yanjiang West Road, Guangzhou Guangdong Province, 510120, China.
| |
Collapse
|
3
|
Micucci M, Xiang BZ, Ting CM, Kwan HY, Mari M, Retini M, Burattini S, Osman R, Okeke UJ, Abdullah FO, Gianfanti F, Battistelli M. Matching traditional Chinese medicine and western medicine-based research: Advanced nutraceutical development for proactive gastric cancer prevention. World J Gastrointest Oncol 2024; 16:3798-3819. [PMID: 39350981 PMCID: PMC11438774 DOI: 10.4251/wjgo.v16.i9.3798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/26/2024] [Accepted: 07/24/2024] [Indexed: 09/09/2024] Open
Abstract
Gastric cancer (GC), the third leading cause of cancer-related death globally, is complex and heterogeneous. This review explores multidisciplinary investigations of traditional Chinese medicine (TCM) combined with Western medical practices, emphasizing the development of nutraceuticals for cancer prevention. Using advanced analytical chemistry and food chemistry techniques, this study investigated how TCM components may be optimized for nutraceutical development. Focusing on molecular interactions with GC pathways, particularly the NF-κB, PI3K/Akt, and Wnt/β-catenin pathways, we examined the effects of TCM polyherbal formulas, extracts, and isolated compounds. These agents modulate apoptosis and cellular proliferation, underscoring their potential in preventive strategies. The convergence of nutraceutical and medicine food homology studies highlights a significant shift towards integrating TCM-derived compounds in a preventive health framework. This approach aims not only to enhance efficacy and reduce side effects but also to champion a preventive paradigm using personalized medicine to advance proactive health maintenance and disease prevention. The combination of TCM and western medical practices offers promising avenues for future research and practical applications in GC prevention.
Collapse
Affiliation(s)
- Matteo Micucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Bian-Zhao Xiang
- Hong Kong Chinese Medicine Clinical Study Centre, Chinese EQUATOR Centre, School of Chinese Medicine, Chinese Clinical Trial Registry (Hong Kong), Hong Kong Baptist University, Hong Kong 999077, China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong 999077, China
| | - Chen-Min Ting
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Hiu-Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Sabrina Burattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Riham Osman
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Udodinma Jude Okeke
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| | - Fuad Othman Abdullah
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil 44001, Iraq
- Department of Pharmacognosy, Faculty Pharmacy, Tishk International University, Erbil 44001, Iraq
| | - Federico Gianfanti
- Institute of Oncology Research, Bellinzona CH6500, Switzerland
- Università della Svizzera Italiana, Lugano CH6900, Switzerland
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino 61029, Italy
| |
Collapse
|
4
|
Yin C, Lan T, Wu Y, Cai J, Li H, Kuang X, Jiao L, Ou X, Yang H, Liu B, Lu W. Integrating Network Pharmacology and Experimental Validation to Investigate the Mechanism of Qushi Huatan Decoction Against Coronary Heart Disease. Drug Des Devel Ther 2024; 18:4033-4049. [PMID: 39280256 PMCID: PMC11402371 DOI: 10.2147/dddt.s463054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/17/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose This study was designed to evaluate the effect and mechanism of the Qushi Huatan (QSHT) decoction against coronary heart disease (CHD) through network pharmacology and experimental verification. Methods In the present study, the active ingredients of the QSHT decoction were identified by ultra performance liquid chromatography/tandem mass spectrometry (UPLC/MS), then the potential ingredients and coronary heart disease targets were predicted using the SwissTarget Prediction database and the database of Genecards and OMIM database, respectively. A herb-compound-target network was constructed using Cytoscape. GO and KEGG enrichment analysis were performed using the ClusterProfiler data package of R software. Molecular docking was used to predict the core targets of QSHT against CHD. In addition, we used a myocardial infarction (MI) and high-fat diet ApoE-/- mice model to investigate the cardioprotective effects of QSHT. Western blotting and immunochemistry were used to verify the core targets and the signaling pathway. Results A total of 68 active ingredients were found in the QSHT decoction. Network pharmacology indicated 28 targets and 147 signal pathways, including AKT1, HIF-1α, GSK-3β, TLR4 and NF-κB, those key targets were also verified by molecular docking. The results of GO and KEGG enrichment analysis showed that the targets of QSHT against CHD were largely associated with inflammatory and oxidative stress, and AKT/HIF-1α and TLR4/NF-κB pathways might be key functional pathways. In vivo, QSHT significantly improved cardiac function and attenuated fibrosis and inflammation. Furthermore, QSHT could significantly inhibit the expression of HIF-1α, TLR4, phosphorylation of AKT1, GSK-3β and NF-κB after MI in ApoE-/- mice. Conclusion Based on network pharmacology, molecular docking and experimental verification, this study demonstrated that QSHT could improve cardiac function and attenuate cardiac fibrosis by regulating TLR4/NF-κB and AKT/HIF-1α signaling pathway in post- MI and high-fat diet ApoE-/- mice.
Collapse
Affiliation(s)
- Chunxia Yin
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Taohua Lan
- Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, People's Republic of China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yunshan Wu
- Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Jing Cai
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Haoxiang Li
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Xiaolan Kuang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Lin Jiao
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Xiaomin Ou
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Hua Yang
- Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, People's Republic of China
| | - Bo Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Weihui Lu
- Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, People's Republic of China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, People's Republic of China
| |
Collapse
|
5
|
Yang L, Liu J, Zhang J, Shao F, Jin Y, Xing J, Zhou H, Yu A. Anticancer effects of Erzhimaoling decoction in high-grade serous ovarian cancer in vitro and in vivo. Eur J Med Res 2024; 29:405. [PMID: 39103890 PMCID: PMC11299366 DOI: 10.1186/s40001-024-01968-4] [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: 05/29/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) is a common gynecologic malignancy with a poor prognosis. The traditional Chinese medicine formula Erzhimaoling decoction (EZMLD) has anticancer potential. This study aims to elucidate the anticancer effects of EZMLD on HGSOC in vitro and in vivo. MATERIALS AND METHODS EZMLD-containing serum was prepared from Sprague-Dawley rats for treating SKOV3 ovarian cancer cells at varying concentrations for 24 h and 48 h to determine the IC50. Concentrations of 0%, 5%, and 10% for 24 h were chosen for subsequent in vitro experiments. The roles of METTL3 and METTL14 in SKOV3 cells were explored by overexpressing these genes and combining EZMLD with METTL3/14 knockdown. Investigations focused on cell viability and apoptosis, apoptosis-related protein expression, and KRT8 mRNA m6A modification. For in vivo studies, 36 BALB/c nude mice were divided into six groups involving EZMLD (6.75, 13.5, and 27 g/kg) and METTL3 or METTL14 knockdowns, with daily EZMLD gavage for two weeks. RESULTS In vitro, EZMLD-containing serum had IC50 values of 8.29% at 24 h and 5.95% at 48 h in SKOV3 cells. EZMLD-containing serum decreased SKOV3 cell viability and increased apoptosis. EZMLD upregulated METTL3/14 and FAS-mediated apoptosis proteins, while downregulating Keratin 8 (KRT8). EZMLD increased KRT8 mRNA m6A methylation. METTL3/14 overexpression reduced SKOV3 cell viability and increased apoptosis, while METTL3/14 knockdown mitigated EZMLD's effects. In vivo, EZMLD suppressed SKOV3 xenografts growth, causing significant apoptosis and modulating protein expression. CONCLUSIONS EZMLD has therapeutic potential for ovarian cancer and may be considered for other cancer types. Future research may explore its broader effects beyond cell apoptosis.
Collapse
MESH Headings
- Female
- Animals
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/genetics
- Humans
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Mice
- Apoptosis/drug effects
- Mice, Nude
- Mice, Inbred BALB C
- Rats
- Cell Proliferation/drug effects
- Rats, Sprague-Dawley
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/genetics
- Methyltransferases/genetics
- Methyltransferases/metabolism
- Cell Survival/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Li Yang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Jingfang Liu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Jiejie Zhang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Feng Shao
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Yanlu Jin
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Jie Xing
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China
| | - Heran Zhou
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Stadium Road, Hangzhou, 310007, Zhejiang, China.
| | - Aijun Yu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
6
|
Guo D, Xu K, Wan Q, Yu S, Ma C, Zhang B, Liu Y, Qu L. Different processing methods and pharmacological effects of Atractylodis Rhizoma. Chin J Nat Med 2024; 22:756-768. [PMID: 39197965 DOI: 10.1016/s1875-5364(24)60591-1] [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: 02/13/2024] [Indexed: 09/01/2024]
Abstract
Atractylodis Rhizoma, a traditional Chinese medicine with an extensive history of treating gastrointestinal disorders and other diseases, undergoes various processing methods in China to enhance its therapeutic efficacy for specific conditions. However, a comprehensive report detailing the changes in chemical composition and pharmacological effects due to these processing methods is currently lacking. This article provides a systematic review of the commonly employed processing techniques for Atractylodis Rhizoma, including raw Atractylodis Rhizoma (SCZ), bran-fried Atractylodis Rhizoma (FCZ), deep-fried Atractylodis Rhizoma (JCZ), and rice water-processed Atractylodis Rhizoma (MCZ). It examines the alterations in chemical constituents and pharmacological activities resulting from these processes and elucidates the mechanisms of action of the primary components in the various processed forms of Atractylodis Rhizoma in the treatment of gastrointestinal diseases.
Collapse
Affiliation(s)
- Dongmei Guo
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Kang Xu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Qianyun Wan
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Songyang Yu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Chaoyang Ma
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Baohui Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China.
| | - Linghang Qu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430065, China.
| |
Collapse
|
7
|
Chen SH, Wu HS, Jiang XF, Zhou C, Bian XR, He X, Li B, Dong YJ, Wang KG, Shen SH, Lv GY, Zhi YH. Bioinformatics and LC-QTOF-MS based discovery of pharmacodynamic and Q-markers of Pitongshu against functional dyspepsia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118096. [PMID: 38537841 DOI: 10.1016/j.jep.2024.118096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pitongshu (PTS) is a clinically effective empirical formula for the treatment of FD. The efficacy and safety of PTS have been demonstrated in randomized, controlled, double-blind trials, but there is a lack of understanding of the systematic evaluation of the efficacy of PTS and its material basis. OBJECTIVE To investigate the efficacy of PTS in Functional dyspepsia (FD) mice and possible Q-markers. METHOD In this study, we used "irregular feeding + chronic unpredictable chronic stimulation" to establish a mice model of FD with hepatogastric disharmony. The efficacy of PTS was assessed from hair condition, behavioral, pain, gastrointestinal function, and serum 5-HT, GAS, MTL levels in mice by instillation of different doses of PTS. In addition, the composition of drugs in blood was analyzed by LC-QTOF-MS and potential Q-markers were selected by combining network pharmacology, molecular docking and actual content. RESULT Our study showed that different doses of PTS increased pain threshold and writhing latency, decreased the number of writhings, increased gastric emptying rate and small intestinal propulsion rate, decreased total acidity of gastric contents and gastric acid secretion, and increased serum levels of 5-HT, GAS, and MTL in mice to different degrees. Enrichment analysis showed that PTS may be anti-FD through multiple pathways such as Serotonergic synapse, thyroid hormone signaling pathway, cholinergic synapse, and dopaminergic synapse. In addition, potential active ingredient substances were explored by LC-QTOF-MS combined with bioinformatics. Combined with the actual contentselected six constituents, hesperidin, neohesperidin, naringin, paeoniflorin, magnolol and honokiol, possible as Q-markers. CONCLUSION PTS may exert its anti-FD effects through multi-component, multi-target and multi-pathway". Constituents, hesperidin, neohesperidin, naringin, paeoniflorin, magnolol and honokiol may be the Q-markers of its anti-FD effects.
Collapse
Affiliation(s)
- Su-Hong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Han-Song Wu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Xiao-Feng Jiang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Cong Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Xue-Ren Bian
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Xinglishang He
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Ying-Jie Dong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, No. 18, Chaowang Road, Xiacheng District, Hangzhou, Zhejiang, 310014, China; College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District, Hangzhou, Zhejiang, 310014, China; Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, zhejiang 313200, China
| | - Kun-Gen Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310006, China; Kun-Gen Wang National Famous Chinese Medicine Doctor Studio, Hangzhou, Zhejiang, 310006, China.
| | - Shu-Hua Shen
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310006, China; Kun-Gen Wang National Famous Chinese Medicine Doctor Studio, Hangzhou, Zhejiang, 310006, China.
| | - Gui-Yuan Lv
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District, Hangzhou, Zhejiang, 310014, China.
| | - Yi-Hui Zhi
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310006, China; Kun-Gen Wang National Famous Chinese Medicine Doctor Studio, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
8
|
Zheng X, Liu J, Hu W, Jiang B, Zhou X, Zhang M, Song M. Curcumin Induces Autophagy-mediated Ferroptosis by Targeting the PI3K/AKT/mTOR Signaling Pathway in Gastric Cancer. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35. [PMID: 39150386 PMCID: PMC11363205 DOI: 10.5152/tjg.2024.23526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/27/2024] [Indexed: 08/17/2024]
Abstract
As a very common malignancy of the digestive system, the incidence and mortality rates of gastric cancer (GC) are increasing year by year. The critical role of ferroptosis in cancer development has been well-documented. The polyphenol compound curcumin shows prominent anti-tumor effects in multiple cancer types, including GC. However, whether curcumin participates in GC tumorigenesis by regulating ferroptosis remains unknown. Gastric cancer cells AGS and HGC-27 were treated with curcumin (0, 10, and 20 μM). Cell viability and death were evaluated through CCK-8 and LDH release assays. LC3B expression in cells was estimated through immunofluorescence staining. Intracellular ferrous iron (Fe2+), GSH, MDA, and lipid ROS levels were assessed by corresponding assay kits. The cellular levels of autophagy markers (ATG5, ATG7, Beclin 1, and LC3B), ferroptosis markers (ACSL4, SLC7A11, and GPX4), and phosphorylated (p)-PI3K, p-AKT, and p-mTOR were determined through western blotting. Curcumin attenuated cell viability but stimulated cell death in GC cells. Curcumin enhanced autophagy in GC cells, as demonstrated by the increased levels of ATG5, ATG7, Beclin 1, and LC3B. Besides, curcumin upregulated iron, MDA, GSH, and ACSL4 levels while downregulated lipid ROS, SLC7A11, and GPX4 levels, suggesting its stimulation on ferroptosis in GC cells. Curcumin decreased p-PI3K, p-AKT, and p-mTOR levels in cells. Importantly, the ferroptosis inhibitor ferrostatin-1 overturned the impacts of curcumin on GC cell viability, death, and ferroptosis. Curcumin suppresses GC development by inducing autophagy-mediated ferroptosis by inactivating the PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Xin Zheng
- Department of General Surgery, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Jun Liu
- Department of General Surgery, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Wei Hu
- Department of General Surgery, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Bin Jiang
- Department of General Surgery, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Xin Zhou
- Department of Neurosurgery, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Min Zhang
- Department of General Surgery, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Ming Song
- Department of General Surgery, Wuhan Third Hospital, Wuhan, Hubei, China
| |
Collapse
|
9
|
Stouffer M, Wandling E, Dickson L, Lin S, Duan H, Powe E, Jean‐Louis D, Tiwari AK, Amos S. Gedunin modulates cellular growth and apoptosis in glioblastoma cell lines. Cancer Rep (Hoboken) 2024; 7:e2051. [PMID: 38702989 PMCID: PMC11069102 DOI: 10.1002/cnr2.2051] [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: 11/17/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Glioblastomas are characterized by aggressive behavior. Surgery, radiotherapy, and alkylating agents, including temozolomide are the most common treatment options for glioblastoma. Often, conventional therapies fail to treat these tumors since they develop drug resistance. There is a need for newer agents to combat this deadly tumor. Natural products such as gedunin have shown efficacy in several human diseases. A comprehensive study of gedunin, an heat shock protein (HSP)90 inhibitor, has not been thoroughly investigated in glioblastoma cell lines with different genetic modifications. AIMS A key objective of this study was to determine how gedunin affects the biological and signaling mechanisms in glioblastoma cells, and to determine how those mechanisms affect the proliferation and apoptosis of glioblastoma cells. METHODS The viability potentials of gedunin were tested using MTT, cell counts, and wound healing assays. Gedunin's effects on glioma cells were further validated using LDH and colony formation assays. In addition, we investigated the survival and apoptotic molecular signaling targets perturbed by gedunin using Western blot analysis and flow cytometry. RESULTS Our results show that there was a reduction in cell viability and inhibition of wound healing in the cells tested. Western blot analysis of the gene expression data revealed genes such as EGFR and mTOR/Akt/NF kappa B to be associated with gedunin sensitivity. Gedunin treatment induced apoptosis by cleaving poly ADP-ribose polymerase, activating caspases, and downregulating BCL-xL. Based on these results, gedunin suppressed cell growth and HSP client proteins, resulting in apoptosis in glioblastoma cell lines. CONCLUSION Our data provide in vitro support for the anticancer activity of gedunin in glioma cells by downregulating cancer survival proteins.
Collapse
Affiliation(s)
- Michael Stouffer
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Elizabeth Wandling
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Lindsay Dickson
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Stacy Lin
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Huanyun Duan
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Erika Powe
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Denise Jean‐Louis
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| | - Amit K. Tiwari
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Samson Amos
- Department of Pharmaceutical SciencesCedarville University School of Pharmacy, Cedarville UniversityCedarvilleOhioUSA
| |
Collapse
|
10
|
Gao J, Tan W, Yuan L, Wang H, Wen J, Sun K, Chen X, Wang S, Deng W. Antitumour mechanisms of traditional Chinese medicine elicited by regulating tumour-associated macrophages in solid tumour microenvironments. Heliyon 2024; 10:e27220. [PMID: 38463777 PMCID: PMC10923716 DOI: 10.1016/j.heliyon.2024.e27220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Tumour-associated macrophages (TAMs), particularly M2-TAMs, constitute the largest proportion of immune cells in the solid tumour microenvironment, playing a crucial role in tumour progression and correlating with poor prognosis. TAMs promote the proliferation, invasion, and metastasis of tumour cells by remodelling the extracellular matrix, inhibiting immunity, promoting immune escape and tumour angiogenesis, and affecting cell metabolism. Traditional Chinese medicine (TCM) has been used clinically in China for millennia. Chinese herbs exhibit potent antitumour effects with minimal to no toxicity, substantially contributing to prolonging the lives of patients with cancer and improving their quality of life. TCM has unique advantages in improving the solid tumour microenvironment, particularly in regulating TAMs to further inhibit tumour angiogenesis, reduce drug resistance, reverse immunosuppression, and enhance antitumour immunity. This review highlights the TAM-associated mechanisms within the solid tumour microenvironment, outlines the recent advancements in TCM targeting TAMs for antitumour effects, emphasises the superiority of combining TCM with standard treatments or new nano-drug delivery systems, and evaluates the safety and efficacy of TCM combined with conventional treatments via clinical trials to provide insights and strategies for future research and clinical treatment.
Collapse
Affiliation(s)
- Jiamin Gao
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Weishan Tan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Luyun Yuan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Haoyue Wang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Junkai Wen
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Kexiang Sun
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Xin Chen
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Shuyun Wang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| | - Wanli Deng
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200135, China
| |
Collapse
|
11
|
Tian S, Ren L, Liu C, Wang Z. Atractylenolide II Suppresses Glycolysis and Induces Apoptosis by Blocking the PADI3-ERK Signaling Pathway in Endometrial Cancer Cells. Molecules 2024; 29:939. [PMID: 38474453 PMCID: PMC10934053 DOI: 10.3390/molecules29050939] [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: 01/05/2024] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Atractylenolide II (AT-II), the major bioactive compound of Atractylodes macrocephala, exhibits anti-cancer activity against many types of tumors, but the roles and the potential mechanisms in endometrial cancer remain unclear. In the present study, AT-II treatment was found to significantly suppress RL95-2 and AN3CA cell proliferation and glycolysis, and induced their apoptosis by inactivating the ERK signaling pathway, accompanied by the changing expression of the glycolytic key enzymes and apoptotic-related proteins. Peptidyl arginine deiminase 3 (PADI3), as the candidate target gene of AT-II, was highly expressed in the endometrial cancer tissues and associated with a poor prognosis according to bioinformatics analysis. PADI3 knockdown inhibited proliferation and glycolysis in endometrial cancer cells and induced cell apoptosis. Furthermore, AT-II negatively regulated the expression of PADI3, and PADI3 overexpression reversed the effects of AT-II on endometrial cancer cells. Our findings suggested that the anti-cancer function of AT-II is associated with the suppression of glycolysis and induction of apoptosis by blocking the PADI3-ERK signaling pathway. Thus, AT-II represents a novel therapeutic target for endometrial cancer and targeting AT-II may serve as a potential strategy for the clinical therapy of endometrial cancer.
Collapse
Affiliation(s)
- Shuang Tian
- Department of Pathology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China;
- Department of Cell Biology and Genetics, Basic Medical College, Jinzhou Medical University, Jinzhou 121001, China
| | - Lili Ren
- Department of Neurobiology, Basic Medical College, Jinzhou Medical University, Jinzhou 121001, China;
| | - Chao Liu
- Department of Cell Biology and Genetics, Basic Medical College, Jinzhou Medical University, Jinzhou 121001, China
| | - Zhe Wang
- Department of Pathology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China;
| |
Collapse
|
12
|
Yuan C, Wu S, Wu Y, Tian C, Wang Z, Zhang X. Effects of Traditional Chinese Medicine "Fuzheng Qingdu Decoction" on Autonomic Function and Cancer-Related Symptoms in Patients with Advanced Gastric Cancer undergoing Chemotherapy: A Controlled Trial. Integr Cancer Ther 2024; 23:15347354241229414. [PMID: 38323452 PMCID: PMC10851715 DOI: 10.1177/15347354241229414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/16/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
OBJECTIVE To evaluate the effects of Fuzheng Qingdu Decoction (FZQDD) on the autonomic function and cancer-related symptoms of patients with advanced gastric cancer undergoing chemotherapy to verify its clinical efficacy. METHODS Sixty-two patients with stage III or IV gastric cancer were included in this study. The patients were divided into 2 groups: the chemotherapy (33 patients) and chemotherapy with FZQDD (29 patients) groups. The primary outcome was the autonomic function of the patients before and after the interventions. The parameters that were used to assess autonomic function were deceleration capacity (DC) and acceleration capacity (AC) of heart rate and heart rate variability (HRV), which comprised standard deviation of the normal-normal interval (SDNN), root mean square of successive interval differences (RMSSD), low-frequency power (LF), high-frequency power (HF), total power (TP), and LF-HF ratio. The secondary outcomes were cancer-related symptoms and the quality of life. RESULTS DC and HRV parameters (ie, SDNN, RMSSD, LF, HF, and TP) were significantly decreased in the chemotherapy group; however, AC significantly increased after the interventions. No significant differences were observed in the DC, AC, and HRV parameters before and after the interventions in the chemotherapy with FZQDD group. Nevertheless, the changes in DC, AC, and HRV parameters (SDNN, RMSSD, HF, and TP) before and after the interventions were statistically significant between both the groups. FZQDD significantly improved the cancer-related symptoms and the quality of life of the patients. CONCLUSIONS Oxaliplatin combined with S-1 (tegafur, gimeracil, and oteracil potassium) can impair autonomic modulation in patients with advanced gastric cancer. FZQDD can alleviate autonomic dysfunction by increasing the parasympathetic activity and decreasing the sympathetic tone, helping patients restore the dynamic sympathovagal balance, and significantly improving the cancer-related symptoms and the quality of life of patients.
Collapse
Affiliation(s)
- Chengjia Yuan
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shuang Wu
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Wu
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Cuiling Tian
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zaichuan Wang
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaochun Zhang
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou, Jiangsu, China
| |
Collapse
|
13
|
Xie Z, Lin M, He X, Dong Y, Chen Y, Li B, Chen S, Lv G. Chemical Constitution, Pharmacological Effects and the Underlying Mechanism of Atractylenolides: A Review. Molecules 2023; 28:molecules28103987. [PMID: 37241729 DOI: 10.3390/molecules28103987] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Atractylenolides, comprising atractylenolide I, II, and III, represent the principal bioactive constituents of Atractylodes macrocephala, a traditional Chinese medicine. These compounds exhibit a diverse array of pharmacological properties, including anti-inflammatory, anti-cancer, and organ-protective effects, underscoring their potential for future research and development. Recent investigations have demonstrated that the anti-cancer activity of the three atractylenolides can be attributed to their influence on the JAK2/STAT3 signaling pathway. Additionally, the TLR4/NF-κB, PI3K/Akt, and MAPK signaling pathways primarily mediate the anti-inflammatory effects of these compounds. Atractylenolides can protect multiple organs by modulating oxidative stress, attenuating the inflammatory response, activating anti-apoptotic signaling pathways, and inhibiting cell apoptosis. These protective effects extend to the heart, liver, lung, kidney, stomach, intestine, and nervous system. Consequently, atractylenolides may emerge as clinically relevant multi-organ protective agents in the future. Notably, the pharmacological activities of the three atractylenolides differ. Atractylenolide I and III demonstrate potent anti-inflammatory and organ-protective properties, whereas the effects of atractylenolide II are infrequently reported. This review systematically examines the literature on atractylenolides published in recent years, with a primary emphasis on their pharmacological properties, in order to inform future development and application efforts.
Collapse
Affiliation(s)
- Zhiyi Xie
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Huzhou 313200, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou 313200, China
| | - Minqiu Lin
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Huzhou 313200, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou 313200, China
| | - Xinglishang He
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Huzhou 313200, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou 313200, China
| | - Yingjie Dong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Huzhou 313200, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou 313200, China
| | - Yigong Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Huzhou 313200, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou 313200, China
| | - Suhong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Huzhou 313200, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou 313200, China
| | - Guiyuan Lv
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
14
|
Liu C, Chen H, Zhang Y, Li M, Jiang Q, Wang Z, Yu L, Wang Q, Pan H, Zhuo Y. Combination of chemical profiling and network pharmacology analysis to investigate the potential mechanism of Li-Zhong-Xiao-Pi granules in the treatment of gastric precancerous lesions. Biomed Chromatogr 2023; 37:e5589. [PMID: 36689998 DOI: 10.1002/bmc.5589] [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: 11/23/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023]
Abstract
Li-Zhong-Xiao-Pi granules (LZXP) are effective for treating gastric precancerous lesions (GPL) in traditional Chinese medicine. However, the active compounds of LZXP and their potential therapeutic mechanism in GPL remained unclarified. The purpose of this study is to investigate the chemical composition and potential targets of LZXP. Based on the accurate masses, ion fragments, and literature data, a total of 128 compounds were identified in the LZXP sample using ultra-performance liquid chromatography and quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) in both positive and negative ion modes, and 28 of these compounds were exactly determined by comparison with authentic reference standards. Meanwhile, 11 typical components were quantified via UPLC during a 24 min period. The linearity, accuracy, stability and recovery of the method were all proven. Through the network pharmacological analysis, six chemicals (quercetin, 4'-hydroxywogonin, sinensetin, 5, 7, 8, 3', 4'-pentamethoxyflavanone, 8-gingerdione and quercetin) were identified as the active ingredients, and five LZXP targets (AKT1, CYP1B1, PTGS2, MMP9 and EGFR) were found to be the crucial molecules in the treatment of GPL. This study provides a systematic and applicable method for the rapid screening and identification of the chemical constituents from LZXP, and an effective understanding the mechanism of LZXP in the treatment of GPL.
Collapse
Affiliation(s)
- Chenchen Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiling Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yida Zhang
- Department of Allergy and Clinical Immunology, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiyao Jiang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhendong Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangwen Yu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Zhuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
15
|
Wang C, Jiang H, Peng J, Weng D, Zhang Y, Zhou Y, Zhang Q. Circular RNA circ_SKA3 enhances gastric cancer development by targeting miR-520h. Histol Histopathol 2023; 38:317-328. [PMID: 36134741 DOI: 10.14670/hh-18-521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
PURPOSE To explore the mechanisms of action of circ_SKA3 in gastric cancer (GC), which are still not fully understood. METHODS Subcellular localization assay was used to analyze the localization of circ_SKA3, and Actinomycin D assay was applied to confirm the stability of circ_SKA3. The levels of circ_SKA3, microRNA (miR)-520h, and cell division cycle 42 (CDC42) mRNA were gauged by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels of CDC42 and proliferating cell nuclear antigen (PCNA) were assessed by western blot. Cell proliferation, colony formation, cell cycle distribution, apoptosis, migration, and invasion were detected by 3-[4,5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT), 5-Ethynyl-2'-Deoxyuridine (EdU) incorporation, colony formation, flow cytometry, and transwell assays, respectively. Directed relationship between miR-520h and circ_SKA3 or CDC42 was verified by a dual-luciferase reporter assay. Mouse xenograft experiments were used to elucidate the impact of circ_SKA3 in vivo. RESULTS Overexpression of circ_SKA3 was validated in GC tissues and cells. The down-regulation of circ_SKA3 suppressed proliferation, cell cycle progression, colony formation, migration, invasion, and promoted cell apoptosis in vitro, as well as weakening tumor growth in vivo. Circ_SKA3 directly bound to miR-520h, and circ_SKA3 regulated CDC42 expression through miR-520h. Circ_SKA3 exerted regulatory effects on GC cell behaviors by inhibiting miR-520h. Furthermore, CDC42 was a functional target of miR-520h in regulating GC cell behaviors. CONCLUSION Our findings established a strong molecular mechanism, the miR-520h/CDC42 axis, at least in part, for the oncogenic role of circ_SKA3 in GC.
Collapse
Affiliation(s)
- Chuntao Wang
- Department of Thyroid Gastroenterology and Thoracic Surgery, Suizhou Central Hospital, Suizhou, Hubei, China
| | - Hao Jiang
- Department of Hepatopancreatobiliary Surgery, Suizhou Central Hospital, Suizhou, Hubei, China
| | - Jiaqun Peng
- Department of Thyroid Gastroenterology and Thoracic Surgery, Suizhou Central Hospital, Suizhou, Hubei, China
| | - Duanshun Weng
- Department of Thyroid Gastroenterology and Thoracic Surgery, Suizhou Central Hospital, Suizhou, Hubei, China
| | - Yu Zhang
- Department of Thyroid Gastroenterology and Thoracic Surgery, Suizhou Central Hospital, Suizhou, Hubei, China
| | - Yanxun Zhou
- Department of Gastroenterology, Suizhou Central Hospital, Suizhou, Hubei, China
| | - Qin Zhang
- Department of Thyroid Gastroenterology and Thoracic Surgery, Suizhou Central Hospital, Suizhou, Hubei, China.
| |
Collapse
|
16
|
Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Motahhary M, Saghari S, Sharifi L, Bokaie S, Mirzaei S, Entezari M, Aref AR, Salimimoghadam S, Rashidi M, Taheriazam A, Hushmandi K. STAT3 signaling in prostate cancer progression and therapy resistance: An oncogenic pathway with diverse functions. Biomed Pharmacother 2023; 158:114168. [PMID: 36916439 DOI: 10.1016/j.biopha.2022.114168] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The categorization of cancers demonstrates that prostate cancer is the most common malignancy in men and it causes high death annually. Prostate cancer patients are diagnosed mainly via biomarkers such as PSA test and patients show poor prognosis. Prostate cancer cells rapidly diffuse into different parts of body and their metastasis is also a reason for death. Current therapies for prostate cancer patients include chemotherapy, surgery and radiotherapy as well as targeted therapy. The progression of prostate cancer cells is regulated by different factors that STAT3 signaling is among them. Growth factors and cytokines such as IL-6 can induce STAT3 signaling and it shows carcinogenic impact. Activation of STAT3 signaling occurs in prostate cancer and it promotes malignant behavior of tumor cells. Induction of STAT3 signaling increases glycolysis and proliferation of prostate cancer cells and prevents apoptosis. Furthermore, STAT3 signaling induces EMT mechanism in increasing cancer metastasis. Activation of STAT3 signaling stimulates drug resistance and the limitation of current works is lack of experiment related to role of STAT3 signaling in radio-resistance in prostate tumor. Calcitriol, capsazepine and β-elemonic are among the compounds capable of targeting STAT3 signaling and its inhibition in prostate cancer therapy. In addition to natural products, small molecules targeting STAT3 signaling have been developed in prostate cancer therapy.
Collapse
Affiliation(s)
- Mehrdokht Sadrkhanloo
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sam Saghari
- Department of Health Services Management, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Laleh Sharifi
- Uro-oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saied Bokaie
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc., 6, Tide Street, Boston, MA 02210, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
17
|
Hu J, Cheng M, Li Y, Shi B, He S, Yao Z, Jiang J, Yu H, He Z, Zhao Y, Zheng H, Hua B, Liu R. Ginseng-containing traditional medicine preparations in combination with fluoropyrimidine-based chemotherapy for advanced gastric cancer: A systematic review and meta-analysis. PLoS One 2023; 18:e0284398. [PMID: 37068063 PMCID: PMC10109524 DOI: 10.1371/journal.pone.0284398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND Ginseng-containing traditional medicine preparations (G-TMPs) in combination with fluoropyrimidine-based chemotherapy (FBC) are well-known treatments for advanced gastric cancer (AGC), with a superior efficacy to FBC alone. However, evidence regarding their efficacy remains limited. The purpose of this meta-analysis is to evaluate the efficacy and safety of G-TMPs in combination with FBC for the treatment of AGC. METHODS Eight electronic databases were searched for randomized controlled trials (RCTs) using G-TMPs with FBC for the treatment of AGC. The primary outcome included the tumor response, while the secondary outcomes included the quality of life (QoL), proportions of peripheral blood lymphocytes, adverse drug reactions (ADRs), and levels of cancer biomarkers. The quality of evidence for each outcome was assessed using GRADE profilers. RESULTS A total of 1,960 participants were involved in the 26 RCTs included. Patients treated with FBC plus G-TMPs had better objective response (risk ratio [RR] = 1.23, 95% confidence interval [CI]: 1.13 to 1.35, p < 0.00001) and disease control (RR = 1.13, 95% CI: 1.08 to 1.19, p < 0.00001) rates than those treated with FBC alone. Additionally, the combination group had a better QoL, higher proportions of CD3+ T cells, CD4+ T cells, and natural killer cells, as well as a higher CD4+/CD8+ T-cell ratio. Furthermore, lower levels of CA19-9, CA72-4, and CEA were confirmed in the combination treatment group. In addition, G-TMPs reduced the incidence of ADRs during chemotherapy. CONCLUSION In combination with FBC, G-TMPs can potentially enhance efficacy, reduce ADRs, and improve prognosis for patients with AGC. However, high-quality randomized studies remain warranted. SYSTEMATIC REVIEW REGISTRATION PROSPERO Number: CRD42021264938.
Collapse
Affiliation(s)
- Jiaqi Hu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Mengqi Cheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yue Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Bolun Shi
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shulin He
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ziang Yao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Juling Jiang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huibo Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongning He
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuwei Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Jiang Y, Guo K, Wang P, Zhu Y, Huang J, Ruan S. The antitumor properties of atractylenolides: Molecular mechanisms and signaling pathways. Biomed Pharmacother 2022; 155:113699. [PMID: 36116253 DOI: 10.1016/j.biopha.2022.113699] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/02/2022] Open
Abstract
Drugs that exhibit a high degree of tumor cell selectivity while minimizing normal cell toxicity are an area of active research interest as a means of designing novel antitumor agents. The pharmacological benefits of Chinese herbal medicine-based treatments have been the focus of growing research interest in recent years. Sesquiterpenoids derived from the Atractylodes macrocephala volatile oil preparations exhibit in vitro and in vivo antitumor activity. Atracylenolides exhibit anti-proliferative, anti-metastatic, and immunomodulatory activity in a range of tumor cell lines in addition to being capable of regulating metabolic activity such that it is a promising candidate drug for the treatment of diverse cancers. The present review provides a summary of recent advances in Atractylenolide-focused antitumor research efforts.
Collapse
Affiliation(s)
- Yu Jiang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, China
| | - Kaibo Guo
- Department of Oncology, Affilited Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Peipei Wang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, China
| | - Ying Zhu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, China
| | - Jiaqi Huang
- Department of postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Shanming Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310006, China.
| |
Collapse
|
19
|
Tan Y, Wang H, Xu B, Zhang X, Zhu G, Ge Y, Lu T, Gao R, Li J. Chinese herbal medicine combined with oxaliplatin-based chemotherapy for advanced gastric cancer: A systematic review and meta-analysis of contributions of specific medicinal materials to tumor response. Front Pharmacol 2022; 13:977708. [PMID: 36091754 PMCID: PMC9453215 DOI: 10.3389/fphar.2022.977708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: The incidence and mortality of gastric cancer ranks among the highest, and the 5-year survival rate of advanced gastric cancer (AGC) is less than 10%. Currently, chemotherapy is the main treatment for AGC, and oxaliplatin is an important part of the commonly used chemotherapy regimen for AGC. A large number of RCTs have shown that Chinese herbal medicine (CHM) combined with oxaliplatin-based chemotherapy can improve objective response rate (ORR) and disease control rate (DCR), reduce the toxic and side effects of chemotherapy. There is currently a lack of systematic evaluation of the evidence to account for the efficacy and safety of CHM combined with oxaliplatin-based chemotherapy in AGC. Therefore, we carried out this study and conducted the sensitivity analysis on the herbal composition to explore the potential anti-tumor efficacy. Methods: Databases of PubMed, EMBASE, CENTRAL, Web of Science, the Chinese Biomedical Literature Database, the China National Knowledge Infrastructure, the Wanfang database, and the Chinese Scientific Journals Database were searched from their inception to April 2022. RCTs evaluating the efficacy of CHM combined with oxaliplatin-based chemotherapy on AGC were included. Stata 16 was used for data synthesis, RoB 2 for quality evaluation of included RCTs, and GRADE for quality of synthesized evidence. Additional sensitivity analysis was performed to explore the potential anti-tumor effects of single herbs and combination of herbs. Results: Forty trials involving 3,029 participants were included. Most included RCTs were assessed as "Some concerns" of risk of bias. Meta-analyses showed that compare to oxaliplatin-based chemotherapy alone, that CHM combined with oxaliplatin-based chemotherapy could increase the objective response rate (ORR) by 35% [risk ratio (RR) = 1.35, 95% confidence intervals (CI) (1.25, 1.45)], and disease control rate (DCR) by 12% [RR = 1.12, 95% CI (1.08, 1.16)]. Subgroup analysis showed that compare to SOX, FOLFOX, and XELOX regimens alone, CHM plus SOX, CHM plus FOLFOX, and CHM plus XELOX could significantly increase the ORR and DCR. Sensitivity analysis identified seven herbs of Astragalus, Liquorice, Poria, Largehead Atractylodes, Chinese Angelica, Codonopsis, and Tangerine Peel with potentials to improve tumor response of oxaliplatin-based chemotherapy in AGC. Conclusion: Synthesized evidence showed moderate certainty that CHM plus oxaliplatin-based chemotherapy may promote improvement in tumor response in AGC. CHM treatment is safe for AGC. Due to the poor quality of included RCTs and small samplesizes, the quality of synthesized evidence was not high. Specific combinations of herbs appeared to produce higher contributions to ORR than the herb individually. Each of this seven above mentioned herbs has been shown in experimental studies to potentially contribute to the improvement of tumor response. To support this conclusion, these seven herbs are worthy of further clinical research. Systematic Review Registration: [http://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=262595], identifier [CRD42022262595].
Collapse
Affiliation(s)
- Ying Tan
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Heping Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bowen Xu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yuansha Ge
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Taicheng Lu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ruike Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
20
|
Hispidulin Enhances Temozolomide (TMZ)-Induced Cytotoxicity against Malignant Glioma Cells In Vitro by Inhibiting Autophagy. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:5266770. [PMID: 35800695 PMCID: PMC9256375 DOI: 10.1155/2022/5266770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/28/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022]
Abstract
Temozolomide (TMZ), an oral alkylating agent, is the widely used first-line chemotherapeutic reagent for glioma in clinical practice. However, TMZ-induced autophagy is another cellular process favoring glioma cell survival. This study aimed to explore whether hispidulin can facilitate TMZ-induced cell death of glioma. The MTT assay showed that coadministration with hispidulin and TMZ could significantly decrease the viability of glioma U87MG cells. Meanwhile, hispidulin administration was also observed to promote TMZ-induced apoptosis. Furthermore, additional hispidulin treatment further elevated TMZ-induced expression of Bax, cleaved-caspase-9, and cleaved-caspase-3 protein but decreased Bcl-2 protein expression in U87MG cells. We also observed that hispidulin suppressed TMZ-induced autophagy to promote apoptosis, as showed by decreased AVOs and LC3B-I/II protein expression. These results collectively suggested that the combination of hispidulin and TMZ could improve the antitumor efficiency of TMZ against malignant gliomas.
Collapse
|
21
|
Huang W, Xie W, Zhong H, Cai S, Huang Q, Liu Y, Zeng Z, Liu Y. Cytosolic p53 Inhibits Parkin-Mediated Mitophagy and Promotes Acute Liver Injury Induced by Heat Stroke. Front Immunol 2022; 13:859231. [PMID: 35634298 PMCID: PMC9139682 DOI: 10.3389/fimmu.2022.859231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/20/2022] [Indexed: 11/25/2022] Open
Abstract
Heat stroke (HS) is a severe condition characterized by increased morbidity and high mortality. Acute liver injury (ALI) is a well-documented complication of HS. The tumor suppressor p53 plays an important role in regulation of mitochondrial integrity and mitophagy in several forms of ALI. However, the role of p53-regulated mitophagy in HS-ALI remains unclear. In our study, we discovered the dynamic changes of mitophagy in hepatocytes and demonstrated the protective effects of mitophagy activation on HS-ALI. Pretreatment with 3-MA or Mdivi-1 significantly exacerbated ALI by inhibiting mitophagy in HS-ALI mice. Consistent with the animal HS-ALI model results, silencing Parkin aggravated mitochondrial damage and apoptosis by inhibiting mitophagy in HS-treated normal human liver cell line (LO2 cells). Moreover, we described an increase in the translocation of p53 from the nucleus to the cytoplasm, and cytosolic p53 binds to Parkin in LO2 cells following HS. p53 overexpression using a specific adenovirus or Tenovin-6 exacerbated HS-ALI through Parkin-dependent mitophagy both in vivo and in vitro, whereas inhibition of p53 using siRNA or PFT-α effectively reversed this process. Our results demonstrate that cytosolic p53 binds to Parkin and inhibits mitophagy by preventing Parkin's translocation from the cytosol to the mitochondria, which decreases mitophagy activation and leads to hepatocyte apoptosis in HS-ALI. Overall, pharmacologic induction of mitophagy by inhibiting p53 may be a promising therapeutic approach for HS-ALI treatment.
Collapse
Affiliation(s)
- Wei Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hanhui Zhong
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Zhang Y, Liu Y, Wang J, Jiang Z, Zhang L, Cui Y, Zhao D, Wang Y. Atractylenolide II inhibits tumor-associated macrophages (TAMs)-induced lung cancer cell metastasis. Immunopharmacol Immunotoxicol 2022; 44:227-237. [PMID: 35166628 DOI: 10.1080/08923973.2022.2037629] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE M2-like tumor-associated macrophages (TAMs) play a crucial role in promoting tumor proliferation, angiogenesis, and metastasis. In the current study, we investigated the relationship between macrophage polarization and the antitumor effect of Atractylenolide II (AT-II) in lung cancer cells. MATERIALS AND METHODS Cell viability, migration, and invasion were determined by MTT assay, wound healing assay, and transwell assay, respectively. Flow cytometry analysis showed the percentage of CD206+ cells. Gene expression was determined by real-time PCR, western blotting, and immunofluorescence staining. Lewis lung carcinoma mouse xenograft and metastasis models were used to examine the effects of AT-II on lung cancer in vivo. RESULTS AT-II (2.5 and 5 µM) did not cause significant inhibition of A549 cell viability but markedly inhibited IL-4/IL-13-induced M2-like polarization, evidenced by the decreased expression of the M2 surface marker CD206, down-regulation of specific M2-marker genes (Arg-1, IL-10 and TGF-β) as well as inhibition of M2 macrophages-mediated invasion and migration of A549 cells. In addition, AT-II inhibited IL-4/IL-13-induced activation of the STAT6 signaling pathway that is vital in the M2-like polarization of macrophages. In animal models, administration of AT-II (50 mg kg-1, i.g., QD for 21 days) significantly inhibited tumor growth, reduced pulmonary metastatic nodules, and down-regulated the percentages of M2 macrophages (F4/80+ and CD206+) in total macrophages (F4/80+) in tumor tissues and pulmonary metastatic nodules. CONCLUSIONS AT-II effectively inhibits M2-like polarization, thereby inhibiting lung cancer cell metastasis both in vivo and in vitro, revealing a novel potential strategy for the antitumor effect of AT-II.
Collapse
Affiliation(s)
- Yunting Zhang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Yuxi Liu
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Jianguang Wang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Zongying Jiang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Lin Zhang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Yong Cui
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Danyu Zhao
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Yanjie Wang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| |
Collapse
|
23
|
Zhang D, Li X, Song D, Chen S, Zhang Z, Cao S, Liu M. Atractylenolide III induces apoptosis by regulating the Bax/Bcl-2 signaling pathway in human colorectal cancer HCT-116 Cells in vitro and in vivo. Anticancer Drugs 2022; 33:30-47. [PMID: 34261915 DOI: 10.1097/cad.0000000000001136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Atractylodes is the dry root of atractylodes macrocephala koidz and has been commonly used as a traditional Chinese medicine (TCM). Atractylenolide III, a main component of atractylodes, has displayed significant effects on anti-inflammation and anticancer. However, the effects of atractylenolide III on growth inhibition and apoptosis induction in colon cancer remain unclear. The results showed that atractylenolide III significantly inhibited the cell growth and induce cellular apoptosis in HCT-116 cells in a concentration dependence manner in vitro. Mechanistic studies further showed that atractylenolide III could regulate the Bax/Bcl-2 apoptotic signaling pathway through promoting the expression of proapoptotic related gene/proteins Bax, caspase-9 and caspase-3 but inhibiting the expression of antiapoptotic related gene/protein Bcl-2 in HCT-116 cells. Furthermore, atractylenolide III also significantly inhibited the tumor growth of HCT-116 tumor xenografts bearing in nude mice through inducing apoptosis by upregulation of the expressions of Bax, cleaved caspase-3 and p53 but downregulation of the expressions of Bcl-2 in HCT-116 tumor tissues in vivo. The studies may provide the scientific rationale for the understanding of the anticancer effect of atractylenolide III. Therefore, atractylenolide III may have the potential to be developed as a promising novel anticancer agent for the treatment of colorectal cancer clinically.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University
- Pharmaceutical Department of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University
| | - Xiaofang Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Daqiang Song
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Siwei Chen
- Pharmaceutical Department of Traditional Chinese Medicine, School of Pharmacy, Southwest Medical University
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University
| | - Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University
| |
Collapse
|
24
|
Zhou W, Zhang H, Wang X, Kang J, Guo W, Zhou L, Liu H, Wang M, Jia R, Du X, Wang W, Zhang B, Li S. Network pharmacology to unveil the mechanism of Moluodan in the treatment of chronic atrophic gastritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153837. [PMID: 34883416 DOI: 10.1016/j.phymed.2021.153837] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/08/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Moluodan (MLD) is a traditional Chinese patent medicine for the treatment of chronic atrophic gastritis (CAG). However, the mechanism of action (MoA) of MLD for treating CAG still remain unclear. PURPOSE Elucidate the MoA of MLD for treating CAG based on network pharmacology. STUDY DESIGN Integrate computational prediction and experimental validation based on network pharmacology. METHODS Computationally, compounds of MLD were scanned by LC-MS/MS and the target profiles of compounds were identified based on network-based target prediction method. Compounds in MLD were compared with western drugs used for gastritis by hierarchical clustering of target profile. Key biological functional modules of MLD were analyzed, and herb-biological functional module network was constructed to elucidate combinatorial rules of MLD herbs for CAG. Experimentally, MLD's effect on different biological functional modules were validated from both phenotypic level and molecular level in 1- Methyl-3-nitro-1-nitrosoguanidine (MNNG)-induced GES-1 cells. RESULTS Computational results show that the target profiles of compounds in MLD can cover most of the biomolecules reported in literature. The MoA of MLD can cover most types of MoA of western drugs for CAG. The treatment of CAG by MLD involved the regulation of various biological functional modules, e.g., inflammation/immune, cell proliferation, cell apoptosis, cell differentiation, digestion and metabolism. Experimental results show that MLD can inhibit cell proliferation, promote cell apoptosis and differentiation, reduce the inflammation level and promote lipid droplet accumulation in MNNG-induced GES-1 cells. CONCLUSION The network pharmacology framework integrating computational prediction and experimental validation provides a novel way for exploring the MoA of MLD.
Collapse
Affiliation(s)
- Wuai Zhou
- Institute of TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Huan Zhang
- TCM Network Pharmacology Department, Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Xin Wang
- Institute of TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jun Kang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Wuyan Guo
- TCM Network Pharmacology Department, Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Lihua Zhou
- TCM Network Pharmacology Department, Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China; College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Huiyun Liu
- Hebei (Handan) TCM Industrial Technology Research Institute, Handan Pharmaceutical Co., Ltd., Handan 056000, China
| | - Menglei Wang
- Hebei (Handan) TCM Industrial Technology Research Institute, Handan Pharmaceutical Co., Ltd., Handan 056000, China
| | - Ruikang Jia
- Hebei (Handan) TCM Industrial Technology Research Institute, Handan Pharmaceutical Co., Ltd., Handan 056000, China
| | - Xinjun Du
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Weihua Wang
- Center of Pharmaceutical Technology, Tsinghua University, China
| | - Bo Zhang
- TCM Network Pharmacology Department, Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China.
| | - Shao Li
- Institute of TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
25
|
Deng M, Chen H, Long J, Song J, Xie L, Li X. Atractylenolides (I, II, and III): a review of their pharmacology and pharmacokinetics. Arch Pharm Res 2021; 44:633-654. [PMID: 34269984 DOI: 10.1007/s12272-021-01342-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 07/08/2021] [Indexed: 02/05/2023]
Abstract
Atractylodes macrocephala Koidz is a widely used as a traditional Chinese medicine. Atractylenolides (-I, -II, and -III) are a class of lactone compounds derived from Atractylodes macrocephala Koidz. Research into atractylenolides over the past two decades has shown that atractylenolides have anti-cancer, anti-inflammatory, anti-platelet, anti-osteoporosis, and antibacterial activity; protect the nervous system; and regulate blood glucose and lipids. Because of structural differences, both atractylenolide-I and atractylenolide-II have remarkable anti-cancer activities, and atractylenolide-I and atractylenolide-III have remarkable anti-inflammatory and neuroprotective activities. We therefore recommend further clinical research on the anti-cancer, anti-inflammatory and neuroprotective effects of atractylenolides, determine their therapeutic effects, alone or in combination. To investigate their ability to regulate blood glucose and lipid, as well as their anti-platelet, anti-osteoporosis, and antibacterial activities, both in vitro and in vivo studies are necessary. Atractylenolides are rapidly absorbed but slowly metabolized; thus, solubilization studies may not be necessary. However, due to the inhibitory effects of atractylenolides on metabolic enzymes, it is necessary to pay attention to the possible side effects of combining atractylenolides with other drugs, in clinical application. In short, atractylenolides have considerable medicinal value and warrant further study.
Collapse
Affiliation(s)
- Mao Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Huijuan Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Jiaying Long
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Jiawen Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, P. R. China.
| |
Collapse
|
26
|
Zhang WJ, Zhao ZY, Chang LK, Cao Y, Wang S, Kang CZ, Wang HY, Zhou L, Huang LQ, Guo LP. Atractylodis Rhizoma: A review of its traditional uses, phytochemistry, pharmacology, toxicology and quality control. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113415. [PMID: 32987126 PMCID: PMC7521906 DOI: 10.1016/j.jep.2020.113415] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 09/04/2020] [Accepted: 09/20/2020] [Indexed: 05/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atractylodis Rhizoma (AR), mainly includes Atractylodes lancea (Thunb.) DC. (A. lancea) and Atractylodes chinensis (DC.) Koidz. (A. chinensis) is widely used in East Asia as a diuretic and stomachic drug, for the treatment of rheumatic diseases, digestive disorders, night blindness, and influenza as it contains a variety of sesquiterpenoids and other components of medicinal importance. AIM OF THE REVIEW A systematic summary on the botany, traditional uses, phytochemistry, pharmacology, toxicology, and quality control of AR was presented to explore the future therapeutic potential and scientific potential of this plant. MATERIALS AND METHODS A review of the literature was performed by consulting scientific databases including Google Scholar, Web of Science, Baidu Scholar, Springer, PubMed, ScienceDirect, CNKI, etc. Plant taxonomy was confirmed to the database "The Plant List". RESULTS Over 200 chemical compounds have been isolated from AR, notably sesquiterpenoids and alkynes. Various pharmacological activities have been demonstrated, especially improving gastrointestinal function and thus allowed to assert most of the traditional uses of AR. CONCLUSIONS The researches on AR are extensive, but gaps still remain. The molecular mechanism, structure-activity relationship, potential synergistic and antagonistic effects of these components need to be further elucidated. It is suggested that further studies should be carried out in the aspects of comprehensive evaluation of the quality of medicinal materials, understanding of the "effective forms" and "additive effects" of the pharmacodynamic substances based on the same pharmacophore of TCM, and its long-term toxicity in vivo and clinical efficacy.
Collapse
Affiliation(s)
- Wen-Jin Zhang
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China; College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen-Yu Zhao
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Li-Kun Chang
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ye Cao
- Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Sheng Wang
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chuan-Zhi Kang
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hong-Yang Wang
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Li Zhou
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lu-Qi Huang
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Lan-Ping Guo
- State Key Laboratory of Dao-di Herbs Breeding Base, Joint Laboratory of Infinitus (China) Herbs Quality Research, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
27
|
Bailly C. Atractylenolides, essential components of Atractylodes-based traditional herbal medicines: Antioxidant, anti-inflammatory and anticancer properties. Eur J Pharmacol 2020; 891:173735. [PMID: 33220271 DOI: 10.1016/j.ejphar.2020.173735] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 12/20/2022]
Abstract
The rhizome of the plant Atractylodes macrocephala Koidz is the major constituent of the Traditional Chinese Medicine Baizhu, frequently used to treat gastro-intestinal diseases. Many traditional medicine prescriptions based on Baizhu and the similar preparation Cangzhu are used in China, Korea and Japan as Qi-booster. These preparations contain atractylenolides, a small group of sesquiterpenoids endowed with antioxidant and anti-inflammatory properties. Atractylenolides I, II and III also display significant anticancer properties, reviewed here. The capacity of AT-I/II/IIII to inhibit cell proliferation and to induce cancer cell death have been analyzed, together with their effects of angiogenesis, metastasis, cell differentiation and stemness. The immune-modulatory properties of ATs are discussed. AT-I has been tested clinically for the treatment of cancer-induced cachexia with encouraging results. ATs, alone or combined with cytotoxic drugs, could be useful to treat cancers or to reduce side effects of radio and chemotherapy. Several signaling pathways have been implicated in their multi-targeted mechanisms of action, in particular those involving the central regulators TLR4, NFκB and Nrf2. A drug-induced reduction of inflammatory cytokines production (TNFα, IL-6) also characterizes these molecules which are generally weakly cytotoxic and well tolerated in vivo. Inhibition of Janus kinases (notably JAK2 and JAK3 targeted by AT-I and AT-III, respectively) has been postulated. Information about their metabolism and toxicity are limited but the long-established traditional use of the Atractylodes and the diversity of anticancer effects reported with AT-I and AT-III should encourage further studies with these molecules and structurally related natural products.
Collapse
|
28
|
Jun X, Fu P, Lei Y, Cheng P. Pharmacological effects of medicinal components of Atractylodes lancea (Thunb.) DC. Chin Med 2018; 13:59. [PMID: 30505341 PMCID: PMC6260578 DOI: 10.1186/s13020-018-0216-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/20/2018] [Indexed: 12/22/2022] Open
Abstract
Atractylodes lancea Thunb. DC. (AL) has a long history as one of the important herbs used in East Asia. This review is on the purpose of providing a comprehensive summary of the pharmacological effects of AL and its extractions. The publication from PubMed, ScienceDirect, Springer, and Wiley database was collected and summarized. The potential application of AL on the disease could be attributed to its pharmacological properties such as anti-cancer, anti-inflammatory and other essential effects. Hence, this review aims at providing evidence of the pharmacological activities of AL as one of natural products used in clinical trial.
Collapse
Affiliation(s)
- Xie Jun
- 1Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Fu
- 2West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yu Lei
- 1Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Cheng
- 1Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|