1
|
Mo N, Zhou P, Liu F, Su H, Han L, Lu C. Integrating network pharmacology, molecular docking, and experimental validation to reveal the mechanism of Radix Rehmanniae in psoriasis. Medicine (Baltimore) 2024; 103:e40211. [PMID: 39470475 PMCID: PMC11520997 DOI: 10.1097/md.0000000000040211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Radix Rehmanniae (RR) plays an important role in treating psoriasis. However, the active compounds of RR and potential mechanisms are unclear. The current study was designed to investigate the potential active ingredients, targets, and mechanisms of RR in treating psoriasis through network pharmacology, molecular docking, and vitro experiments. METHODS Initially, the TCMSP database and literature retrieval were used to access the active ingredients of RR. The psoriasis target proteins were obtained from Therapeutic Target Database, OMIM, GeneCards, and DrugBank databases. The target proteins were then converted into target genes using Uniprot. Secondly, overlapping genes were obtained through Venn online tool. Then, protein-protein interactions network diagram is finished by STRING database. Next, Cytoscape software was used to acquire the top 10 hub proteins; gene ontology and Kyoto encyclopedia of genes and genomes enrichment analysis were then used to predict possible mechanisms. Afterwards, molecular docking validation of the active ingredients with the main targets was performed by AutoDock software. Finally, lipopolysaccharides induced RAW264.7, to assess the effects and molecular mechanisms by MTT, RT-qPCR, and Western blot assays. RESULTS Overall, there are 20 effective compounds and 33 targets involved in biological processes including apoptosis, intracellular signaling, vasodilation, and mitogen-activated protein kinase (MAPK) signaling cascade. The docking results showed strong binding capacity between the active ingredients and targets. We verified aucubin as the key active ingredient, tumor necrosis factor α, and IL6 as the core targets, and focused on the p38MAPK protein pathway. Cellular experiments showed that aucubin down-regulated the phosphorylated p38MAP protein and reduced the expression of tumor necrosis factor α mRNA, IL6 mRNA, and IL1βmRNA. CONCLUSION In summary, RR is featured with multicomponent, multi-target, and multi-pathway in treating psoriasis; the preliminary mechanism may be associated with the down-regulation of p38MAPK phosphorylation and curbing the expression of inflammatory factor by aucubin. This paper provides the scientific basis for Traditional Chinese medicine treating psoriasis.
Collapse
Affiliation(s)
- Nian Mo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Panyu Zhou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fanlu Liu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haojie Su
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Han
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of TCM Moisture Syndrome at the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of TCM and Immune Disease Research, Guangzhou, China
- Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
| | - Chuanjian Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of TCM Moisture Syndrome at the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of TCM and Immune Disease Research, Guangzhou, China
- Guangdong Province Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Liu S, Teng F, Lu Y, Zhu Y, Liang X, Wu F, Liu J, Zhou W, Su C, Cao Y. Ethoxy-erianin phosphate inhibits angiogenesis in colorectal cancer by regulating the TMPO-AS1/miR-126-3p/PIK3R2 axis and inactivating the PI3k/AKT signaling pathway. BMC Cancer 2024; 24:1275. [PMID: 39402462 PMCID: PMC11476319 DOI: 10.1186/s12885-024-12893-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy, with increasing prevalence and mortality. How the ethoxy-erianin phosphate (EBTP) mediates CRC development remains unclear. Therefore, the current study evaluated the effects of EBTP on the proliferation, migration, and angiogenesis of CRC cells using CCK-8, Wound-healing, Transwell, and Tube formation assays. RNA sequencing and molecular docking techniques helped predict that EBTP could inhibit angiogenesis by regulating PIK3R2 expression while clarifying the mechanism behind EBTP-mediated CRC angiogenesis. Subsequently, several in vitro experiments indicated that PIK3R2 overexpression significantly improved the proliferation, migration, and angiogenesis of CRC cells while knocking down PIK3R2 expression inhibited their proliferation, migration, and angiogenesis. Simultaneously, PIK3R2 expression in CRC cells gradually decreased with increased EBTP concentration and action duration. Moreover, PIK3R2 overexpression in CRC cells could reverse the inhibitory EBTP effect in angiogenesis. Mouse experiments also depicted that EBTP inhibited CRC angiogenesis by down-regulating PIK3R2 expression. In addition, EBTP could inhibit PI3K/AKT pathway activity and indirectly control PIK3R2 expression through the lncRNA TMPO-AS1/miR-126-3p axis. Our findings highlighted that EBTP could inhibit CRC angiogenesis using the TMPO-AS1/miR-126-3p/PIK3R2/PI3k/AKT axis, providing a novel strategy for anti-angiogenic therapy in CRC.
Collapse
Affiliation(s)
- Shaoqun Liu
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai, China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Fei Teng
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai, China
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China
| | - Yuxin Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yanqing Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xin Liang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Fanhong Wu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, China
- Zhejiang Guangsha Vocational and Technical University of Construction, Jinhua, China
| | - Jianwen Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
- Zhejiang Guangsha Vocational and Technical University of Construction, Jinhua, China
| | - Wenming Zhou
- Department of Endoscope Center, Minhang Hospital, Fudan University, Shanghai, China.
| | - Chang Su
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China.
| | - Yiou Cao
- Department of Gastrointestinal Surgery, Minhang Hospital, Fudan University, Shanghai, China.
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Ni JR, Zhang QH, Deng JL, Wang HH, Duan YC, Zhang CJ, Jiang LT. Promotion Effect of Catalpol on Angiogenesis and Potential Mechanisms: A Research Based on Network Pharmacology. Chem Biol Drug Des 2024; 104:e14602. [PMID: 39134897 DOI: 10.1111/cbdd.14602] [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: 10/20/2023] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 08/18/2024]
Abstract
Catalpol, a natural iridoid glycoside, has potential therapeutic benefits, including anti-inflammatory and neuroprotective effects. Investigating catalpol's role in angiogenesis is critical for understanding its potential therapeutic applications, particularly in diseases where modulating angiogenesis is beneficial. This study investigates catalpol's influence on angiogenesis and its mechanisms, combining network pharmacology and in vitro experiments. The target genes corresponding to the catalpol were analyzed by SwissTargetPrediction. Then angiogenesis-related targets were acquired from databases like GeneCards. Subsequently, the Database for Annotation, Visualization and Integrated Discovery was employed for Gene Ontology and pathway analysis, while Cytoscape visualized protein interactions. The effect of catalpol on viability and angiogenesis of HUVECs was further examined using Cell Counting Kit-8 and angiogenesis assays. RT-qPCR and western blot were applied to check the expression of angiogenesis-related proteins. Totally, 312 target genes of catalpol and 823 angiogenesis-related targets were obtained with 56 common targets leading to PPI network analysis, highlighting hub genes (AKT1, EGFR, STAT3, MAPK3, and CASP3). These hub genes were mainly enriched in lipid and atherosclerosis pathway and EGFR-related pathway. The in vitro experimental results showed that catalpol achieved a concentration-dependent increase in HUVECs viability. Catalpol also promoted the migration and angiogenesis of HUVECs and up-regulated the expression of EGFR. EGFR knockdown inhibited the effect of catalpol on HUVECs. Catalpol promotes angiogenesis in HUVECs by upregulating EGFR and angiogenesis-related proteins, indicating its potential therapeutic application in vascular-related diseases.
Collapse
Affiliation(s)
- Jin-Rong Ni
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Qun-Hu Zhang
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Jie-Lin Deng
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Hai-Hu Wang
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Yong-Chi Duan
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Cheng-Ji Zhang
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Lue-Tao Jiang
- Department of Orthopedics, Jiangsu Province (Suqian) Hospital, Suqian, China
| |
Collapse
|
4
|
Yan H, Wang X, Yu L, Liu X, Yan F, Xie Y, Pu Q, Yang Z. Effectiveness of adjuvant traditional Chinese medicine on macrovascular invasion in patients with hepatocellular carcinoma: a real-world propensity score-matched study. Front Pharmacol 2024; 15:1353720. [PMID: 38464727 PMCID: PMC10923102 DOI: 10.3389/fphar.2024.1353720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
The study aimed to investigate the potential of traditional Chinese medicine (TCM) in reducing the risk of macrovascular invasion (MVI) in Chinese patients with hepatocellular carcinoma (HCC). This retrospective analysis involved 2,267 HCC patients treated at our hospital. Propensity score (PS) matching was used to compare TCM users (n = 485) with non-users (n = 485) in terms of age, Barcelona Clinic Liver Cancer (BCLC) staging, type of treatment, and AFP. The impact of TCM on the hazard ratio (HR) of MVI was evaluated using a Cox multivariate regression model. The efficacy of TCM therapy on MVI was further examined using the log-rank test. The analysis revealed that TCM medication was a significant protective factor for MVI in HCC patients, as evidenced by the Cox analysis (adjusted HR = 0.496, 95% CI: 0.387-0.635, p < 0.001). After PS matching, the Kaplan-Meier curve demonstrated a lower occurrence rate of MVI in TCM users compared to non-users. The study findings suggest that TCM treatment has the potential to decrease the incidence of MVI in HCC patients, irrespective of etiology, BCLC staging, liver function, or treatment type. Notably, as the use of TCM increased, the percentage of MVI in patients showed a gradual decrease, indicating the potential of TCM therapy as a successful strategy for preventing MVI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhiyun Yang
- Center of Integrative Medicine, Beijing Ditan Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Lu C, Zhang S, Lei SS, Wang D, Peng B, Shi R, Chong CM, Zhong Z, Wang Y. A comprehensive review of the classical prescription Yiguan Jian: Phytochemistry, quality control, clinical applications, pharmacology, and safety profile. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117230. [PMID: 37778517 DOI: 10.1016/j.jep.2023.117230] [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: 06/19/2023] [Revised: 09/10/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yiguan Jian (YGJ) is a classical prescription, which employs 6 kinds of medicinal herbs including Rehmanniae Radix, Lycii Fructus, Angelicae sinensis Radix, Glehniae Radix, Ophiopogonis Radix, and Toosendan Fructus. YGJ decoction is originally prescribed in Qing Dynasty (1636 CE ∼ 1912 CE) in China, and is commonly used to treat liver diseases. There remain abundant literature investigating YGJ decoction from multiple aspects, but few reviews summarized the research and gave a precise definition, which impedes further applications and commercialization of YGJ decoction. AIM OF THE REVIEW The aim of this review is to provide comprehensive descriptions of YGJ decoction, tackling with issues in the research and development of YGJ decoction. MATERIALS AND METHODS The literature and clinical reports were obtained from the databases including Web of Science, Science Direct, PubMed, Google Scholar, China National Knowledge Infrastructure, China Science Periodical Database, China Science and Technology Journal Database, and SinoMed since 2000. The phytochemical characteristics, quality control, pharmaceutical forms, clinical position, pharmacological effects, and toxic events of YGJ decoction were included for analysis. RESULT This review firstly summarized the progress of the chemical existences of YGJ decoction and discussed the advanced methods in monitoring quality of YGJ decoction and its herbal ingredients, particularly in the form of granules. Whilst this review aims to identify the pharmacological actions and clinical impacts of YGJ decoction, the medicinal materials that could provide these benefits were observed in the remaining herbs to exert the anti-fibrotic effects, anti-inflammatory activities, anti-cancer, and anti-diabetic effects, and to universally treat liver and gastric diseases. This review provided supplementary descriptions on the safety issues, especially in Glehniae Radix and Toosendan Fructus, to define the alterations between hepatoprotective activities and unclear toxics in YGJ decoction application. CONCLUSIONS Our comprehensively organized review discussed the chemical characteristics and the research in altering or identifying these essences. The effects of YGJ decoction on the non-clinical and clinical tests exert the good management of sophisticated diseases. In this review, current issues are discussed to inform and inspire subsequent research of YGJ decoction and other classical prescriptions.
Collapse
Affiliation(s)
- Changcheng Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Siyuan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Si San Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Danni Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Bo Peng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Ruipeng Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Cheong-Meng Chong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
6
|
Li Y, Zhai X, Ma L, Zhao L, An N, Feng W, Huang L, Zheng X. Transcriptome Analysis Provides Insights into Catalpol Biosynthesis in the Medicinal Plant Rehmannia glutinosa and the Functional Characterization of RgGES Genes. Genes (Basel) 2024; 15:155. [PMID: 38397145 PMCID: PMC10888080 DOI: 10.3390/genes15020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
Rehmannia glutinosa, a member of the Scrophulariaceae family, has been widely used in traditional Chinese medicine since ancient times. The main bioactive component of R. glutinosa is catalpol. However, the biogenesis of catalpol, especially its downstream pathway, remains unclear. To identify candidate genes involved in the biosynthesis of catalpol, transcriptomes were constructed from R. glutinosa using the young leaves of three cultivars, Beijing No. 3, Huaifeng, and Jin No. 9, as well as the tuberous roots and adventitious roots of the Jin No. 9 cultivar. As a result, 71,142 unigenes with functional annotations were generated. A comparative analysis of the R. glutinosa transcriptomes identified over 200 unigenes of 13 enzymes potentially involved in the downstream steps of catalpol formation, including 9 genes encoding UGTs, 13 for aldehyde dehydrogenases, 70 for oxidoreductases, 44 for CYP450s, 22 for dehydratases, 30 for decarboxylases, 19 for hydroxylases, and 10 for epoxidases. Moreover, two novel genes encoding geraniol synthase (RgGES), which is the first committed enzyme in catalpol production, were cloned from R. glutinosa. The purified recombinant proteins of RgGESs effectively converted GPP to geraniol. This study is the first to discover putative genes coding the tailoring enzymes mentioned above in catalpol biosynthesis, and functionally characterize the enzyme-coding gene in this pathway in R. glutinosa. The results enrich genetic resources for engineering the biosynthetic pathway of catalpol and iridoids.
Collapse
Affiliation(s)
- Yuanjun Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| | - Xiaoru Zhai
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| | - Ligang Ma
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| | - Le Zhao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| | - Na An
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| | - Longyu Huang
- National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
- State Key Laboratory of Cotton Biology, Institute of Cotton Research of Chinese Academy of Agricultural Sciences, Anyang 455000, China
- Hainan Yazhou Bay Seed Laboratory, Sanya 572024, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (Y.L.)
| |
Collapse
|
7
|
Jia J, Chen J, Wang G, Li M, Zheng Q, Li D. Progress of research into the pharmacological effect and clinical application of the traditional Chinese medicine Rehmanniae Radix. Biomed Pharmacother 2023; 168:115809. [PMID: 37907043 DOI: 10.1016/j.biopha.2023.115809] [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: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023] Open
Abstract
The traditional Chinese medicine (TCM) Rehmanniae Radix (RR) refers to the fresh or dried root tuber of the plant Rehmannia glutinosa Libosch of the family Scrophulariaceae. As a traditional Chinese herbal medicine (CHM), it possesses multiple effects, including analgesia, sedation, anti-inflammation, antioxidation, anti-tumor, immunomodulation, cardiovascular and cerebrovascular regulation, and nerve damage repair, and it has been widely used in clinical practice. In recent years, scientists have extensively studied the active components and pharmacological effects of RR. Active ingredients mainly include iridoid glycosides (such as catalpol and aucuboside), phenylpropanoid glycosides (such as acteoside), other saccharides, and unsaturated fatty acids. In addition, the Chinese patent medicine (CPM) and Chinese decoction related to RR have also become major research subjects for TCM practitioners; one example is the Bolus of Six Drugs, which includes Rehmannia, Lily Bulb and Rehmannia Decoction, and Siwu Decoction. This article reviews recent literature on RR; summarizes the studies on its chemical constituents, pharmacological effects, and clinical applications; and analyzes the progress and limitations of current investigations to provide reference for further exploration and development of RR.
Collapse
Affiliation(s)
- Jinhao Jia
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China
| | - Jianfei Chen
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China
| | - Guoli Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China
| | - Minjing Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China
| | - Qiusheng Zheng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003 Xinjiang, PR China.
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832003 Xinjiang, PR China.
| |
Collapse
|
8
|
Liu J, Liu S, Yu M, Li J, Xie Z, Gao B, Liu Y. Anti-inflammatory effect and mechanism of catalpol in various inflammatory diseases. Drug Dev Res 2023; 84:1376-1394. [PMID: 37534768 DOI: 10.1002/ddr.22096] [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: 04/10/2023] [Revised: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
Catalpol is a kind of iridoid glucoside, widely found in a variety of plants, mostly extracted from the rhizome of the traditional medicinal herb rehmanniae. It has various biological activities such as anti-inflammatory, antioxidant, and antitumor. The anti-inflammatory effects of catalpol have been demonstrated in a variety of diseases, such as neurological diseases, atherosclerosis, renal diseases, respiratory diseases, digestive diseases, bone and joint diseases, eye diseases, and periodontitis. The purpose of this review is to summarize the existing literature on the anti-inflammatory effects of catalpol in a variety of inflammatory diseases over the last decade and to focus on the anti-inflammatory mechanisms of catalpol.
Collapse
Affiliation(s)
- Jinyao Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shuang Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Mingyue Yu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jianing Li
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zunxuan Xie
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Boyang Gao
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuyan Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
9
|
Qiu FS, Wang JF, Guo MY, Li XJ, Shi CY, Wu F, Zhang HH, Ying HZ, Yu CH. Rgl-exomiR-7972, a novel plant exosomal microRNA derived from fresh Rehmanniae Radix, ameliorated lipopolysaccharide-induced acute lung injury and gut dysbiosis. Biomed Pharmacother 2023; 165:115007. [PMID: 37327587 DOI: 10.1016/j.biopha.2023.115007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/01/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023] Open
Abstract
Plant-derived exosome-like nanoparticles (ELNs) have been proposed as a novel therapeutic tool for preventing human diseases. However, the number of well-verified plant ELNs remains limited. In this study, the microRNAs in ELNs derived from fresh Rehmanniae Radix, a well-known traditional Chinese herb for treating inflammatory and metabolic diseases, were determined by using microRNA sequencing to investigate the active components in the ELNs and the protection against lipopolysaccharide (LPS)-induced acute lung inflammation in vivo and in vitro. The results showed that rgl-miR-7972 (miR-7972) was the main ingredient in ELNs. It exerted stronger protective activities against LPS-induced acute lung inflammation than catalpol and acteoside, which are two well-known chemical markers in this herb. Moreover, miR-7972 decreased the production of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), reactive oxygen species (ROS) and nitric oxide (NO) in LPS-exposed RAW264.7 cells, thereby facilitating M2 macrophage polarization. Mechanically, miR-7972 downregulated the expression of G protein-coupled receptor 161 (GPR161), activating the Hedgehog pathway, and inhibited the biofilm form of Escherichia coli via targeting virulence gene sxt2. Therefore, miR-7972 derived from fresh R. Radix alleviated LPS-induced lung inflammation by targeting the GPR161-mediated Hedgehog pathway, recovering gut microbiota dysbiosis. It also provided a new direction for gaining novel bioactivity nucleic acid drugs and broadening the knowledge on cross-kingdom physiological regulation through miRNAs.
Collapse
Affiliation(s)
- Fen-Sheng Qiu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China
| | - Jia-Feng Wang
- Department of Pharmacy, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mei-Ying Guo
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China
| | - Xue-Jian Li
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China
| | - Chang-Yi Shi
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China; Westlake University, Hangzhou 310024, China
| | - Fang Wu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China
| | - Huan-Huan Zhang
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China
| | - Hua-Zhong Ying
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China.
| | - Chen-Huan Yu
- Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences (Hangzhou Medical College), Hangzhou Medical College, Hangzhou 310013, China; Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310022, China; Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou 310018, China.
| |
Collapse
|
10
|
Xie ZY, Cao HW, Wang Q, Lu H, Du W. Catalpol inhibits hepatic stellate cell activation by reducing the formation and changing the contents of hepatocyte-derived extracellular vesicles. J Cell Commun Signal 2023; 17:723-736. [PMID: 36508052 PMCID: PMC10409968 DOI: 10.1007/s12079-022-00716-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Hepatic stellate cell (HSC) activation is the central event in hepatic fibrosis. The cross-talk between HSCs and hepatocytes, which is mediated by extracellular vesicles (EVs), affects HSC activation. This study aimed to investigate whether Catalpol (CTP) attenuated hepatic fibrosis via modulating EVs. Mice were injected intraperitoneally with CCl4 for 4 weeks to induce hepatic fibrosis. They were gavaged with CTP daily. Mouse serum EVs were isolated and identified using nanoparticle tracking analysis and transmission electron microscopy. Mouse hepatocytes (AML12) and primary HSCs were used to investigate the cell-to-cell crosstalk. The autophagosome-autolysosome fusion was determined using the autophagic flux assay. Hepatic fibrosis was attenuated by CTP, with a decrease of the myofibroblast marker, alpha-smooth muscle actin. The CTP treatment lowered the serum EVs. The co-culture of HSCs and the EVs derived from the CTP-treated mice or hepatocytes reduced HSC proliferation and the expressions of ACTA2 and Col1a1. After the CCl4 treatment, the autophagosomes in AML12 cells were increased, while the autolysosomes were reduced. The decrease of autophagic cargo receptor SQSTM1 in the CTP group suggested that autophagic degradation was sustained. After inhibiting the endogenous Rac1-GTP of hepatocytes, the co-culture of EVs and HSCs reduced Rac1-GTP. The Rac1-GTP level in serum EVs from the CTP-treated mice was reduced in vivo. CTP inhibited autophagy in hepatocytes by reducing Rac1-GTP and thus affect the amount of Rac1-GTP in hepatocyte-derived EVs and the formation of EVs, which attenuated hepatic fibrosis via inhibiting HSC activation.
Collapse
Affiliation(s)
- Zheng-Yuan Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No.1 Mingde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| | - Heng-Wei Cao
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Qing Wang
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Hui Lu
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Wen Du
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
11
|
Li S, Tian Z, Xian X, Yan C, Li Q, Li N, Xu X, Hou X, Zhang X, Yang Y, Xue S, Ma S, Cui S, Sun L, Yao X. Catalpol rescues cognitive deficits by attenuating amyloid β plaques and neuroinflammation. Biomed Pharmacother 2023; 165:115026. [PMID: 37336148 DOI: 10.1016/j.biopha.2023.115026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/04/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023] Open
Abstract
This study sought to investigate the anti-amyloid β (Aβ) and anti-neuroinflammatory effects of catalpol in an Alzheimer's disease (AD) mouse model. METHODS The effects of catalpol on Aβ formation were investigated by thioflavin T assay. The effect of catalpol on generating inflammatory cytokines from microglial cells and the cytotoxicity of microglial cells on HT22 hippocampal cells were assessed by real-time quantitative PCR, ELISA, redox reactions, and cell viability. APPswe/PS1ΔE9 mice were treated with catalpol, and their cognitive ability was investigated using the water maze and novel object recognition tests. Immunohistochemistry and immunofluorescence were used to probe for protein markers of microglia and astrocyte, Aβ deposits, and NF-κB pathway activity. Aβ peptides, neuroinflammation, and nitric oxide production were examined using ELISA and redox reactions. RESULTS Catalpol potently inhibited Aβ fibril and oligomer formation. In microglial cells stimulated by Aβ, catalpol alleviated the expression of the proinflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and inducible nitric oxide synthase (iNOS) but promoted the expression of the anti-inflammatory cytokine IL-10. Catalpol alleviated the cytotoxic effects of Aβ-exposed microglia on HT22 cells. Treatment with catalpol in APPswe/PS1ΔE9 mice downregulated neuroinflammation production, decreased Aβ deposits in the brains and alleviated cognitive impairment. Catalpol treatment decreased the number of IBA-positive microglia and GFAP-positive astrocytes and their activities of the NF-κB pathway in the hippocampus of APPswe/PS1ΔE9 mice. CONCLUSION The administration of catalpol protected neurons by preventing neuroinflammation and Aβ deposits in an AD mouse model. Therefore, catalpol may be a promising strategy for treating AD.
Collapse
Affiliation(s)
- Si Li
- Department of Technology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Ziqi Tian
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaohui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Cuihuan Yan
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qiang Li
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Nan Li
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaokang Xu
- College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaojie Hou
- College of Acupuncture and Massage, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaoyun Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yinan Yang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Sisi Xue
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shengkai Ma
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shuanlong Cui
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Lijun Sun
- Department of Nephrology, The First Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Xiaoguang Yao
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, College of Integrative Medicine, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| |
Collapse
|
12
|
Kong Y, Liu S, Wang S, Yang B, He W, Li H, Yang S, Wang G, Dong C. Design, synthesis and anticancer activities evaluation of novel pyrazole modified catalpol derivatives. Sci Rep 2023; 13:7756. [PMID: 37173367 PMCID: PMC10182059 DOI: 10.1038/s41598-023-33403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
Catalpol, a natural product mainly existed in plenty of Chinese traditional medicines, is an iridoid compound with the comprehensive effects on neuroprotective, anti-inflammatory, choleretic, hypoglycemic and anticancer. However, there are some disadvantages for catalpol such as a short half-life in vivo, low druggability, stingy binding efficiency to target proteins and so on. It is necessary to make structural modification and optimization which enhance its performance on disease treatments and clinic applications. Pyrazole compounds have been reported to have excellent anticancer activities. Based on the previous research foundation of our research group on iridoids and the anticancer activities of catalpol and pyrazole, a series of pyrazole modified catalpol compounds were synthesized by principle of drug combination for serving as potential cancer inhibitors. These derivatives are characterized by 1H NMR, 13C NMR and HRMS. The efficacy of anti-esophageal cancer and anti-pancreatic cancer activities were evaluated by the MTT assay on two esophageal cancer cells Eca-109 and EC-9706, and two pancreatic cancer cells PANC-1, BxPC-3 and normal pancreatic cell line HPDE6-C7, which showed that the compound 3e had strong inhibitory activity against esophageal cancer cells, this providing a theoretical basis for the discovery of catalpol-containing drugs.
Collapse
Affiliation(s)
- Yuanfang Kong
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Shuanglin Liu
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
- Henan Polysaccharide Research Center, Zhengzhou, 450046, Henan, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, 450046, Henan, China
| | - Shaopei Wang
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
- Henan Polysaccharide Research Center, Zhengzhou, 450046, Henan, China
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, 450046, Henan, China
| | - Bin Yang
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Wei He
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Hehe Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Siqi Yang
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| | - Guoqing Wang
- Department of Applied Chemistry, Zhengzhou University of Light Industry, ZhengzhouHenan, 450001, China.
| | - Chunhong Dong
- Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
- Henan Polysaccharide Research Center, Zhengzhou, 450046, Henan, China.
- Henan Key Laboratory of Chinese Medicine for Polysaccharides and Drugs Research, Zhengzhou, 450046, Henan, China.
| |
Collapse
|
13
|
Ardizzone A, Bova V, Casili G, Repici A, Lanza M, Giuffrida R, Colarossi C, Mare M, Cuzzocrea S, Esposito E, Paterniti I. Role of Basic Fibroblast Growth Factor in Cancer: Biological Activity, Targeted Therapies, and Prognostic Value. Cells 2023; 12:cells12071002. [PMID: 37048074 PMCID: PMC10093572 DOI: 10.3390/cells12071002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer is the leading cause of death worldwide; thus, it is necessary to find successful strategies. Several growth factors, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF, FGF2), and transforming growth factor beta (TGF-β), are involved in the main processes that fuel tumor growth, i.e., cell proliferation, angiogenesis, and metastasis, by activating important signaling pathways, including PLC-γ/PI3/Ca2+ signaling, leading to PKC activation. Here, we focused on bFGF, which, when secreted by tumor cells, mediates several signal transductions and plays an influential role in tumor cells and in the development of chemoresistance. The biological mechanism of bFGF is shown by its interaction with its four receptor subtypes: fibroblast growth factor receptor (FGFR) 1, FGFR2, FGFR3, and FGFR4. The bFGF–FGFR interaction stimulates tumor cell proliferation and invasion, resulting in an upregulation of pro-inflammatory and anti-apoptotic tumor cell proteins. Considering the involvement of the bFGF/FGFR axis in oncogenesis, preclinical and clinical studies have been conducted to develop new therapeutic strategies, alone and/or in combination, aimed at intervening on the bFGF/FGFR axis. Therefore, this review aimed to comprehensively examine the biological mechanisms underlying bFGF in the tumor microenvironment, the different anticancer therapies currently available that target the FGFRs, and the prognostic value of bFGF.
Collapse
Affiliation(s)
- Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Valentina Bova
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | | | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy
| | - Marzia Mare
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
- Correspondence: ; Tel.: +39-090-6765208
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy
| |
Collapse
|
14
|
Ndongwe T, Witika BA, Mncwangi NP, Poka MS, Skosana PP, Demana PH, Summers B, Siwe-Noundou X. Iridoid Derivatives as Anticancer Agents: An Updated Review from 1970-2022. Cancers (Basel) 2023; 15:770. [PMID: 36765728 PMCID: PMC9913650 DOI: 10.3390/cancers15030770] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
The rise of cancer cases has coincided with the urgent need for the development of potent chemical entities and/or modification of existing commodities to improve their efficacy. Increasing evidence suggests that cancer remains one of the leading causes of death globally, with colon cancer cases alone likely to rise exponentially by 2030. The exponential rise in cancer prevalence is largely attributable to the growing change toward a sedentary lifestyle and modern diets, which include genetically modified foods. At present, the prominent treatments for cancer are chemotherapy, surgery, and radiation. Despite slowing cancer progression, these treatments are known to have devastating side effects that may deteriorate the health of the patient, thus, have a low risk-benefit ratio. In addition, many cancer drugs have low bioavailability, thereby limiting their therapeutic effects in cancer patients. Moreover, the drastic rise in the resistance of neoplastic cells to chemotherapeutic agents is rendering the use of some drugs ineffective, thereby signaling the need for more anticancer chemical entities. As a result, the use of natural derivatives as anticancer agents is gaining considerable attention. Iridoids have the potential to form conjugates with other anticancer, antidiabetic, antileishmanial, and antimalarial drugs, which synergistically have the potential to increase their effects. Published studies have identified the role of iridoids, which, if fully explored, may result in cheaper and less toxic alternative/adjuvant cancer drugs. The subject of this article is natural and synthetic iridoid derivatives and their potential therapeutic roles as anticancer agents.
Collapse
Affiliation(s)
- Tanaka Ndongwe
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Bwalya A. Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Nontobeko P. Mncwangi
- Department of Pharmacy Practice, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Madan S. Poka
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Phumzile P. Skosana
- Department of Clinical Pharmacy, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Patrick H. Demana
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Beverley Summers
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| | - Xavier Siwe-Noundou
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Medunsa 0204, South Africa
| |
Collapse
|
15
|
Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural Products as Anticancer Agents: Current Status and Future Perspectives. Molecules 2022; 27:molecules27238367. [PMID: 36500466 PMCID: PMC9737905 DOI: 10.3390/molecules27238367] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Natural products have been an invaluable and useful source of anticancer agents over the years. Several compounds have been synthesized from natural products by modifying their structures or by using naturally occurring compounds as building blocks in the synthesis of these compounds for various purposes in different fields, such as biology, medicine, and engineering. Multiple modern and costly treatments have been applied to combat cancer and limit its lethality, but the results are not significantly refreshing. Natural products, which are a significant source of new therapeutic drugs, are currently being investigated as potential cytotoxic agents and have shown a positive trend in preclinical research and have prompted numerous innovative strategies in order to combat cancer and expedite the clinical research. Natural products are becoming increasingly important for drug discovery due to their high molecular diversity and novel biofunctionality. Furthermore, natural products can provide superior efficacy and safety due to their unique molecular properties. The objective of the current review is to provide an overview of the emergence of natural products for the treatment and prevention of cancer, such as chemosensitizers, immunotherapeutics, combinatorial therapies with other anticancer drugs, novel formulations of natural products, and the molecular mechanisms underlying their anticancer properties.
Collapse
Affiliation(s)
- Abid Naeem
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Zhang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yali Liu
- Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang Medical College, Nanchang 330006, China
- Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang Medical College, Nanchang 330006, China
| | - Weifeng Zhu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Correspondence:
| |
Collapse
|
16
|
Zheng Z, Bian C, Wang H, Su J, Meng L, Xin Y, Jiang X. Prediction of immunotherapy efficacy and immunomodulatory role of hypoxia in colorectal cancer. Ther Adv Med Oncol 2022; 14:17588359221138383. [PMID: 36425871 PMCID: PMC9679351 DOI: 10.1177/17588359221138383] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/26/2022] [Indexed: 11/26/2023] Open
Abstract
Immunotherapy has been used in the clinical treatment of colorectal cancer (CRC); however, most patients fail to achieve satisfactory survival benefits. Biomarkers with high specificity and sensitivity are being increasingly developed to predict the efficacy of CRC immunotherapy. In addition to DNA alteration markers, such as microsatellite instability/mismatch repair and tumor mutational burden, immune cell infiltration and immune checkpoints (ICs), epigenetic changes and no-coding RNA, and gut microbiomes all show potential predictive ability. Recently, the hypoxic tumor microenvironment (TME) has been identified as a key factor mediating CRC immune evasion and resistance to treatment. Hypoxia-inducible factor-1α is the central transcription factor in the hypoxia response that drives the expression of a vast number of survival genes by binding to the hypoxia response element in cancer and immune cells in the TME. Hypoxia regulates angiogenesis, immune cell infiltration and activation, expression of ICs, and secretion of various immune molecules in the TME and is closely associated with the immunotherapeutic efficacy of CRC. Currently, various agents targeting hypoxia have been found to improve the TME and enhance the efficacy of immunotherapy. We reviewed current markers commonly used in CRC to predict therapeutic efficacy and the mechanisms underlying hypoxia-induced angiogenesis and tumor immune evasion. Exploring the mechanisms by which hypoxia affects the TME will assist the discovery of new immunotherapeutic predictive biomarkers and development of more effective combinations of agents targeting hypoxia and immunotherapy.
Collapse
Affiliation(s)
- Zhuangzhuang Zheng
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Chenbin Bian
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Huanhuan Wang
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Jing Su
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Xin Jiang
- Department of Radiation Oncology, the First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| |
Collapse
|
17
|
Cao M, Wang Y, Lu G, Qi H, Li P, Dai X, Lu J. Classical Angiogenic Signaling Pathways and Novel Anti-Angiogenic Strategies for Colorectal Cancer. Curr Issues Mol Biol 2022; 44:4447-4471. [PMID: 36286020 PMCID: PMC9601273 DOI: 10.3390/cimb44100305] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Although productive progress has been made in colorectal cancer (CRC) researchs, CRC is the second most frequent type of malignancy and the major cause of cancer-related death among gastrointestinal cancers. As angiogenesis constitutes an important point in the control of CRC progression and metastasis, understanding the key signaling pathways that regulate CRC angiogenesis is critical in elucidating ways to inhibit CRC. Herein, we comprehensively summarized the angiogenesis-related pathways of CRC, including vascular endothelial growth factor (VEGF), nuclear factor-kappa B (NF-κB), Janus kinase (JAK)/signal transducer and activator of transcription (STAT), Wingless and int-1 (Wnt), and Notch signaling pathways. We divided the factors influencing the specific pathway into promoters and inhibitors. Among these, some drugs or natural compounds that have antiangiogenic effects were emphasized. Furthermore, the interactions of these pathways in angiogenesis were discussed. The current review provides a comprehensive overview of the key signaling pathways that are involved in the angiogenesis of CRC and contributes to the new anti-angiogenic strategies for CRC.
Collapse
Affiliation(s)
- Mengyuan Cao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yunmeng Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guige Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Haoran Qi
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Peiyu Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Correspondence:
| |
Collapse
|
18
|
Wang X, Li S, Yan S, Shan Y, Wang X, Jingbo Z, Wang Y, Shan F, Griffin N, Sun X. Methionine enkephalin inhibits colorectal cancer by remodeling the immune status of the tumor microenvironment. Int Immunopharmacol 2022; 111:109125. [PMID: 35988519 DOI: 10.1016/j.intimp.2022.109125] [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: 06/20/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022]
Abstract
There is evidence that methionine enkephalin (MENK), an opioid peptide, promotes anti-tumor immune responses. In this study, the effect of MENK on colorectal cancer (CRC) and its mechanisms of action were examined in vivo. The intraperitoneal administration of 20 mg/kg MENK effectively inhibited MC38 subcutaneous colorectal tumor growth in mice. MENK inhibited tumor progression by increasing the immunogenicity and recognition of MC38 cells. MENK down-regulated the oncogene Kras and anti-apoptotic Bclxl and Bcl2, suppressed Il1b, Il6, iNOS, and Arg1 (encoding inflammatory cytokines), and increased Il17a and Il10 levels. MENK promoted a tumor suppressive state by decreasing the immune checkpoints Pd-1, Pd-l1, Lag3, Flgl1, and 2b4 in CRC. MENK also altered the immune status of the tumor immune microenvironment (TIME). It increased the infiltration of M1-type macrophages, CD8+T cells, and CD4+T cells and decreased the proportions of G-MDSCs, M-MDSCs, and M2-type macrophages. MENK accelerated CD4+TEM and CD8+TEM cell activation in the TIME and up-regulated IFN-γ, TNF-α, and IL-17A in CD4+T cells and Granzyme B in CD8+T cells. In addition, analyses of PD-1 and PD-L1 expression indicated that MENK promoted the anti-tumor immune response mediated by effector T cells. Finally, OGFr was up-regulated at the protein and mRNA levels by MENK, and the inhibitory effects of MENK on tumor growth were blocked by NTX, a specific blocker of OGFr. These finding indicate that MENK remodels the TIME in CRC to inhibit tumor progression by binding to OGFr. MENK is a potential therapeutic agent for CRC, especially for improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Xiaonan Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| | - Shunlin Li
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Siqi Yan
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yuanye Shan
- Immune Therapeutics Inc., 2431 Aloma Ave #124 Winter Park, FL 32792, USA
| | - Xiao Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| | - Zhai Jingbo
- Medical College, Inner Mongolia Minzu University, Tongliao 028000, China; Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao 028000, China.
| | - Yuanyuan Wang
- Department of Anesthesiology, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Fengping Shan
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| | - Noreen Griffin
- Immune Therapeutics Inc., 2431 Aloma Ave #124 Winter Park, FL 32792, USA
| | - Xun Sun
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
19
|
Huang X, Xu X, Ke H, Pan X, Ai J, Xie R, Lan G, Hu Y, Wu Y. microRNA-16-5p suppresses cell proliferation and angiogenesis in colorectal cancer by negatively regulating forkhead box K1 to block the PI3K/Akt/mTOR pathway. Eur J Histochem 2022; 66. [PMID: 35536149 PMCID: PMC9134092 DOI: 10.4081/ejh.2022.3333] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) have aroused increasing attention in colorectal cancer (CRC) therapy. This study is designed for a detailed analysis of the roles of miR-16-5p and forkhead box K1 (FOXK1) in cell angiogenesis and proliferation during CRC in addition to their underlying mechanisms. CRC tissues and colon cancer cell lines (SW620 and HCT8) were investigated. qRT-PCR and Western blot were utilized to evaluate miR-16-5p and FOXK1 expression. Following gain- and loss-of-function assays on miR-16-5p or FOXK1, the effects of miR-16-5p and FOXK1 were assessed on cell angiogenesis and proliferation in CRC cells. A dual-luciferase reporter assay was employed to evaluate the binding relationship of miR-16-5p and FOXK1. Western blot was used to determine the effects of miR-16-5p and FOXK1 on key molecules of the PI3K/Akt/mTOR pathway. Highly expressed FOXK1 and lowly expressed miR-16-5p were observed in CRC cells and tissues. miR-16-5p overexpression or FOXK1 knockdown reduced CRC cell proliferation and angiogenesis of human umbilical vein endothelial cells co-cultured with the supernatant of CRC cells, whereas miR-16-5p silencing or FOXK1 upregulation caused opposite trends. Additionally, miR-16-5p negatively modulated FOXK1 expression. The blockade of the PI3K/Akt/mTOR pathway was triggered by miR-16-5p overexpression or FOXK1 silencing. In conclusion, miR-16-5p hampers cell angiogenesis and proliferation during CRC by targeting FOXK1 to block the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Xin Huang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Xuan Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Huajing Ke
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Xiaolin Pan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Jiaoyu Ai
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Ruyi Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Guilian Lan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Yang Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| | - Yao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Clinical Research Center for Gastroenterology, Nanchang.
| |
Collapse
|
20
|
Lu MK, Chang CC, Chao CH, Hsu YC. Structural changes, and anti-inflammatory, anti-cancer potential of polysaccharides from multiple processing of Rehmannia glutinosa. Int J Biol Macromol 2022; 206:621-632. [PMID: 35217089 DOI: 10.1016/j.ijbiomac.2022.02.112] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/18/2022] [Accepted: 02/17/2022] [Indexed: 12/31/2022]
Abstract
Polysaccharides play important roles in the bioactivities of Rehmannia glutinosa. This study examined the physiochemical structure and biological activity of the polysaccharides of R. glutinosa during nine steps of processing. Characteristic study showed galactose, glucose, and fructose were the major sugars in the polysaccharides. The percentage of the high-molecular weight polysaccharide increased after processing. In addition, polysaccharides from repeated steam and dry processing of R. glutinosa can effectively increase the anti-inflammatory activity. Secretions of tumor necrosis factor (TNF-α), interleukin (IL)-6, and transforming growth factor (TGF)β after lipopolysaccharide (LPS) stimulation were detected in RAW264.7 macrophages because of its anti-inflammatory activity. RG-B9, a polysaccharide of the ninth steam and dry processing, showed the strongest inhibitory activity on bacterial LPS-induced macrophage IL-6 and TGFβ production. Mechanically, RG-B9 down-regulated the phosphorylation of AKT/ERK. The anti-inflammation of RG-B9 involved AKT/ERK/JNK signaling. In addition, RG-B9 inhibited the viability of lung cancer cells via EGFR/AKT signaling.
Collapse
Affiliation(s)
- Mei-Kuang Lu
- National Research Institute of Chinese Medicine, 155-1 Li-Nung St., Sec. 2, Shipai, Peitou, Taipei 112, Taiwan; Graduate Institute of Pharmacognosy, Taipei Medical University, 252 Wu-Hsing St., Taipei 110, Taiwan.
| | - Chia-Chuan Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Hsein Chao
- National Research Institute of Chinese Medicine, 155-1 Li-Nung St., Sec. 2, Shipai, Peitou, Taipei 112, Taiwan
| | - Yu-Chi Hsu
- National Research Institute of Chinese Medicine, 155-1 Li-Nung St., Sec. 2, Shipai, Peitou, Taipei 112, Taiwan
| |
Collapse
|
21
|
Li M, Jiang H, Hao Y, Du K, Du H, Ma C, Tu H, He Y. A systematic review on botany, processing, application, phytochemistry and pharmacological action of Radix Rehmnniae. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114820. [PMID: 34767834 DOI: 10.1016/j.jep.2021.114820] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Rehmanniae (RR) is the tuber root of Rehmannia glutionsa Libosch, which was firstly recorded in Shennong's Classic of Materia Medica (⟪⟫). RR is a non-toxic and wide used traditional Chinese medicine. RR has the effect of clearing heat, generating essence, cooling blood, stopping bleeding, nourishing yin and blood, and filling marrow. It is used in clinic in the form of processed decoction pieces, including Dry Radix Rehmnniae (DRR) and Rehmanniae Radix Praeparata (RRP). The application of RR in traditional Chinese medicine (TCM) prescriptions can treat various diseases, such as anemia, irregular menstruation, deficiency of liver yin, renal failure and so on. AIM OF REVIEW This paper aims to provide a comprehensive and productive review of RR, which mainly contains botanical characteristics, processing methods, traditional application, chemical composition, quality control and pharmacological action. MATERIALS AND METHODS Literature search was conducted through the Web of Science, Baidu Scholar, ScienceDirect, PubMed, CNKI, and WanFang DATA using the keywords "Radix Rehmnniae", "Rehmanniae Radix Praeparata", "processing", "clinical application", "chemical composition", "quality control", and "pharmacological action". In addition, information was collected from relevant textbooks, reviews, and documents. RESULTS RR is a traditional Chinese herbal medicine with clinical value and rich resources. More than 100 components have been isolated and identified from RR. It has multiple pharmacological actions, such as hemostasis, antioxidation, anti-osteoporosis, lowering blood sugar, improving renal function, anti-inflammation, protecting neuronal function, antidepression and anti-anxiety. DRR and RRP are two different processed products of RR. After processing, there are great changes in property, taste, efficacy, clinical application, chemical composition and pharmacological action. At present, identifying chemical constituents of RR and its medicinal value has been deeply studied. However, there is a lack of research on the reasons for the differences in pharmacological effects between DRR and RRP. The reasons for these differences need to be further verified. Catalpol, the active component of RR, has been studied extensively in the literature, but the pharmacological effects of catalpol cannot represent the pharmacological effects of the whole RR. In the future, effective components such as rehmannioside D, polysaccharide, total glycosides, and effective parts in RR need to be further studied and developed. The pharmacodynamic material basis and mechanism of RR need to be further discussed. The scientific connotation and processing methods of RRP need to be studied and standardized.
Collapse
Affiliation(s)
- Minmin Li
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Huajuan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yule Hao
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Kequn Du
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Hongling Du
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Chuan Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - He Tu
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, 610041, China.
| | - Yao He
- State Key Laboratory of Southwestern Chinese Medicine Resources; Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Guizhou Yibai Pharmaceutical Co. Ltd. Guiyang, 550008, China.
| |
Collapse
|
22
|
Song L, Yin H, Han R, Li J, Ma N, Wang Y, Guo H. Metabolism of Du Zhong Formula in rats using UPLC-Q-TOF/MS. JOURNAL OF MASS SPECTROMETRY : JMS 2022; 57:e4795. [PMID: 34913224 DOI: 10.1002/jms.4795] [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: 06/04/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 06/14/2023]
Abstract
Du Zhong Formula (DZF), a traditional Chinese medicine formula derived from BeiJiQianJinYaoFang, is used to treat kidney deficiency and lumbago. In this study, ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometer (UPLC-Q-TOF/MS) technique combined with pattern recognition analysis was applied for analysis of metabolic profiles of the bioactive components of the DZF in rat biological samples. In this experiment, a total of 73 compounds, including 53 prototype components and 20 metabolites, were identified tentatively in vivo compared with blank urine, plasma, feces, and cerebrospinal fluid (CSF). The prototype ingredients in DZF include terpenoids, gingerols, phenylpropanoids, alkaloids, phenanthrenes, bibenzyls, organic acids, and other ingredients. The metabolic pathways of DZF involved reduction, demethylation, hydroxylation, desugarization, deoxygenation, glucuronidation, sulfation, and methylation. The proposed method could develop an integrated template approach to analyze screening and identification of the bioactive components in plasma, urine, feces, and CSF after oral administration of herb medicines. Additionally, this investigation might provide helpful chemical information for further pharmacology and activity mechanism of DZF.
Collapse
Affiliation(s)
- Lili Song
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongqing Yin
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Han
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingfang Li
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ningning Ma
- School of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hong Guo
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Formula of Traditional Chinese Medicine, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
23
|
Wang G, Yang Y, Yi D, Yuan L, Yin PH, Ke X, Jun-Jie W, Tao MF. Eudragit S100 prepared pH-responsive liposomes-loaded betulinic acid against colorectal cancer in vitro and in vivo. J Liposome Res 2021; 32:250-264. [PMID: 34895013 DOI: 10.1080/08982104.2021.1999974] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This study aimed to develop polymer Eudragit S100 for preparing pH-responsive liposomes-loaded betulinic acid (pH-BA-LP) to improve the therapeutic index of chemotherapy for colorectal cancer. BA-loaded liposomes were coated with Eudragit S100 by a thin film dispersion and easily scalable pH-driven method. The prepared liposomes were evaluated for size, surface morphology, entrapment efficiency, stability, in vitro drug release, and antitumor activity. In particular, pH-BA-LP showed advantages such as lower size (<100 nm), encapsulation efficiency of 90%, high stability, and stably cumulative release. By detecting the antitumor effects of pH-BA-LP in vivo, it showed that the tumor proliferation and cell migration were significantly inhibited in colorectal cancer. The pH-BA-LP also inhibited tumor growth via the regulation of Akt/TLR-mediated signalling and significantly down-regulated the expression of NFAT1 and NFAT4 proteins. It was found that pH-BA-LP can increase NK cells and CD3+ cells in tumor tissues, and the proportion of CD8+ cells in CD3+ cells was also increased, which proved that pH-BA-LP can play an antitumor effect by enhancing the autoimmunity level in tumor-bearing mice. The positive infiltration rates of CD8 and CD68 were increased and CD163 was relatively decreased by using pH-BA-LP, which proved that pH-BA-LP can regulate the immune infiltration levels in tumor-bearing mice. Therefore, the present work provides an effective method to prepare pH-responsive polymer-coated liposomes for colonic delivery with biologically active compounds.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Yu Yang
- Jiangsu University School of Pharmacy, Zhenjiang City, China
| | - Du Yi
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Lu Yuan
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Pei-Hao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Ke
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Jun-Jie
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Min-Fang Tao
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| |
Collapse
|
24
|
Wu Q, Li D, Sun T, Liu J, Ou H, Zheng L, Hou X, Li W, Fan F. Bai-He-Gu-Jin-Tang formula suppresses lung cancer via AKT/GSK3β/β-catenin and induces autophagy via the AMPK/mTORC1/ULK1 signaling pathway. J Cancer 2021; 12:6576-6587. [PMID: 34659548 PMCID: PMC8489124 DOI: 10.7150/jca.62779] [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: 05/15/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022] Open
Abstract
Aims: Bai-He-Gu-Jin-Tang (BHGJT) is a classic Chinese formula used to treat lung cancer, while the underlying molecular mechanism remains obscure. The aim of the study was to investigate the molecular mechanism of BHGJT on lung cancer and demonstrate the potential for synergistic treatment combining BHGJT with conventional therapy. Methods: Cell viability assay, colony formation assay and EdU assay were used to determine the in vitro effects of BHGJT, and a subcutaneous xenograft model was used to evaluate the in vivo effect. Cell cycle analysis, apoptosis rate analysis, immunohistochemical and immunofluorescent staining, Western blot assays and network pharmacology-based analysis were used to explore the underlying mechanisms. Results: We found that BHGJT inhibited cell proliferation via a dose-dependent pathway and obviously hindered tumor growth in vivo in lung cancer. Cell cycle arrest and apoptosis were pronouncedly induced by BHGJT via dysregulation of the cell cycle regulators CDK4 and Cyclin D1 and dysregulation of apoptosis-associated proteins, such as cleaved caspase 3/9 and the BCL-2 family. Based on a network pharmacology-based analysis and experimental evidence, we demonstrated that the AKT/GSK3β/β-catenin signaling pathways were responsible for BHGJT-induced apoptosis in lung cancer cells. Additionally, autophagy was induced by BHGJT via the AMPK/mTORC1/ULK1 signaling pathway, and blocking autophagy with either chloroquine or a ULK1 inhibitor increased the killing efficiency of BHGJT in lung cancer cells. Conclusion: Our findings indicate that the BHGJT formula efficiently inhibits lung cancer growth and represents a potential complementary and alternative treatment for lung cancer.
Collapse
Affiliation(s)
- Quhui Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Da Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Taoli Sun
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, P. R. China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Huiping Ou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Lei Zheng
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xuyang Hou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Fuyuan Fan
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
25
|
Catalpol alleviates Ang II-induced renal injury through NF/κB pathway and TGF-β1/Smads pathway. J Cardiovasc Pharmacol 2021; 79:e116-e121. [PMID: 34654783 DOI: 10.1097/fjc.0000000000001148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Catalpol is an iridoid glycoside obtained from Rehmannia glutinosa, which in previous studies showed various pharmacological properties, including anti-inflammatory, antioxidant, antidiabetic, antitumor and dopaminergic neurons protecting effects. Here, we examined the effect of catalpol on AngII-induced renal injury induced by angiotensin II (AngII), and further to explore its latent molecular mechanisms. We used an in vivo model of AngII-induced renal injury mice, catalpol (25, 50, and 100 mg/kg) was administered for 28 days. Mouse glomerular mesangial cells (SV40 MES 13), rat kidney interstitial fibroblasts cells (NRK-49F), and human proximal tubular epithelial cells (HK-2) were induced by AngII (10 µM) in the presence or absence of catalpol (1, 5, and 10 µM) and incubated for 48 h in vitro. In our study, PAS and masson staining of renal tissue showed that catalpol reduced AngII-induced renal injury in a concentration-dependent manner. The positive expressions of Collagen IV and TGF-β1 were observed to decrease sharply after catalpol treatment. In renal tissue, the levels of pro-inflammatory cytokines TNF-α and IL-6 were evidently decreased after catalpol intervention. Catalpol can relieve AngII-induced renal injury by inactivating NF/κB and TGF-β1/Smads signaling pathways. Therefore, catalpol may act as a potential drug to treat AngII-induced renal injury.
Collapse
|
26
|
El-Hanboshy SM, Helmy MW, Abd-Alhaseeb MM. Catalpol synergistically potentiates the anti-tumour effects of regorafenib against hepatocellular carcinoma via dual inhibition of PI3K/Akt/mTOR/NF-κB and VEGF/VEGFR2 signaling pathways. Mol Biol Rep 2021; 48:7233-7242. [PMID: 34596810 DOI: 10.1007/s11033-021-06715-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/23/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common primary liver cancer characterized by dysregulation of several crucial cellular signaling pathways such as PI3K/p-Akt/mTOR/NF-κB and VEGF/VEGFR2 pathways. Novel therapies targeting these pathways have been discovered such as regorafenib which is small molecular multi-kinase inhibitor mainly targets VEGF/VEGFR2. Catalpol is an iridoid glycoside richly found in rehmannia glutinosa which is a fundamental herb used extensively in traditional Chinese medicine. It is evidenced that catalpol has many pharmacological effects on nervous and cardiovascular systems, in addition to exhibiting hypoglycemic, anti-inflammatory, anti-proliferative and anti-tumour activities. However, its effect on HCC isn't clear enough. So, this study aimed to investigate the anti-tumour effects of catalpol either alone or in combination with regorafenib on HCC. METHODS AND RESULTS In vitro experiments were performed using HepG2 and HUH-7 hepatocellular carcinoma cell lines. MTT assays evaluated anti-proliferative effects of catalpol and/or regorafenib. Combination index was calculated via compusyn software to detect synergism. Tumour biomarkers were measured using ELISA technique. Results showed that catalpol has anti-tumour effects against HCC via targeting PI3K/p-Akt/mTOR/NF-κB and VEGF/VEGFR2 pathways. In addition, results revealed that our novel combination of catalpol and regorafenib showed potent synergistic anti-tumour effect via suppressing both of PI3K/p-Akt/mTOR/NF-κB and VEGF/VEGFR2 signaling pathways and their downstreams. CONCLUSION Catalpol and/or regorafenib markedly suppressed PI3K/p-Akt/mTOR/NF-κB and VEGF/VEGFR2 signaling pathways and consequently showed potent anti-tumour effects against HCC. Results encourage further pre-clinical and clinical studies of this novel combination as a promising targeted therapy for HCC management.
Collapse
Affiliation(s)
- Sara Muhammad El-Hanboshy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, 22514, Egypt.
| | - Maged Wasfy Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, 22514, Egypt
| | | |
Collapse
|
27
|
Huo H, Yang S, Wu H, Sun Y, Zhao R, Ye R, Yan D, Shi X, Yang J. Brain endothelial cells-derived extracellular vesicles overexpressing ECRG4 inhibit glioma proliferation through suppressing inflammation and angiogenesis. J Tissue Eng Regen Med 2021; 15:1162-1171. [PMID: 34551201 DOI: 10.1002/term.3244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/22/2021] [Accepted: 09/02/2021] [Indexed: 12/27/2022]
Abstract
Esophageal cancer related gene-4 (ECRG4) has been shown to be a candidate tumor suppressor in many tumors, but its role in glioma remains poorly understood. This study aimed to explore whether extracellular vesicles (EVs) derived from brain endothelial cells which overexpressed ECRG4 have anti-tumor effect on gliomas in vivo and in vitro, as well as the possible mechanism. A constructed lentivirus expressing the ECRG4 gene was transfected into the hCMEC/D3 cell line. The EVs were isolated from the cells and characterized by Western blot with exosome markers of CD9, CD63, CD81, Alix. RT-PCR and Western blot were performed to verify ECRG4 expression. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and clone formation assays were applied to detect the proliferation of glioma cells incubated with EVs expressing the ECRG4 (ECRG4-exo). The level of inflammatory cytokines and angiogenesis related factors, including nuclear factor kappa-B (NF-κB), interleukin (IL)-1β, IL-6, IL-8, monocyte chemoattractant protein-1 (MCP-1), hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) levels were detected by ELISA. The T98G cell xenograft mouse model was established and treated with ECRG4-EV. The tumor volume and weight were recorded. p38-MAPK, p-p38-MAPK proteins were determined by Western blot in tumor tissues. As a result, EVs can be internalized into U87MG and T98G cells. ECRG4-EV inhibited U87MG and T98G cell proliferation. ECRG4-EV also inhibited the expression of factors involved in inflammation and angiogenesis. In addition, ECRG4-EVs suppressed tumor growth and decreased the production of inflammatory cytokines through inactivation of p38-MAPK signal pathway. In conclusion, ECRG4-EVsuppresses glioma proliferation through modulating the inflammation and angiogenesis.
Collapse
Affiliation(s)
- Haoran Huo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Song Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haotian Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuchen Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ranran Zhao
- Department of Hemodialysis, Traditional Chinese Medicine Hospital of Shi Jia Zhuang City, Shijiazhuang, Hebei, China
| | - Ruihao Ye
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dongdong Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xuefang Shi
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiankai Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
28
|
Potential Roles of Iridoid Glycosides and Their Underlying Mechanisms against Diverse Cancer Growth and Metastasis: Do They Have an Inhibitory Effect on Cancer Progression? Nutrients 2021; 13:nu13092974. [PMID: 34578851 PMCID: PMC8466600 DOI: 10.3390/nu13092974] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Iridoids are glycosides found in plants, having inherent roles in defending them against infection by viruses and microorganisms, and in the rapid repair of damaged areas. The emerging roles of iridoid glycosides on pharmacological properties have aroused the curiosity of many researchers, and studies undertaken indicate that iridoid glycosides exert inhibitory effects in numerous cancers. This review focuses on the roles and the potential mechanism of iridoid glycosides at each stage of cancer development such as proliferation, epithelial mesenchymal transition (EMT), migration, invasion and angiogenesis. Overall, the reviewed literature indicates that iridoid glycosides inhibit cancer growth by inducing cell cycle arrest or by regulating apoptosis-related signaling pathways. In addition, iridoid glycosides suppress the expression and activity of matrix metalloproteinases (MMPs), resulting in reduced cancer cell migration and invasiveness. The antiangiogenic mechanism of iridoid glycosides was found to be closely related to the transcriptional regulation of pro-angiogenic factors, i.e., vascular endothelial growth factors (VEGFs) and cluster of differentiation 31 (CD31). Taken together, these results indicate the therapeutic potential of iridoid glycosides to alleviate or prevent rapid cancer progression and metastasis.
Collapse
|
29
|
Catalpol Weakens Depressive-like Behavior in Mice with Streptozotocin-induced Hyperglycemia via PI3K/AKT/Nrf2/HO-1 Signaling Pathway. Neuroscience 2021; 473:102-118. [PMID: 34358633 DOI: 10.1016/j.neuroscience.2021.07.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022]
Abstract
Depression has huge social risks of high incidence, disability, and suicide. Its prevalence and harm in people with hyperglycemia are 2-3 times higher than in normal people. However, antidepressants with precise curative effects and clear mechanisms for patients with hyperglycemia are currently lacking. Prescriptions containing Rehmannia glutinosa, a traditional medicinal herb with a wide range of nutritional and medicinal values, are often used as antidepressants in Chinese clinical medicine. Catalpol is one of the main effective compounds of R. glutinosa, with multiple biological activities such as hypoglycemia. Here, the antidepressant effect of catalpol on the pathological state of streptozotocin (STZ)-induced hyperglycemia and the underlying molecular mechanisms were analyzed. Results showed that administering catalpol orally to hyperglycemic mice for 21 consecutive days significantly reversed the abnormalities in tail suspension, forced swimming, and open field tests. Catalpol also reversed the abnormal phosphorylation of phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) and the abnormal levels of nuclear factor erythroid 2-related factor 2 (Nrf2) protein, heme oxygenase-1 (HO-1), and antioxidants, including superoxide dismutase, glutathione peroxidase, glutathione-s transferase, reduced glutathione, and malondialdehyde in the hippocampus and frontal cortex of STZ-induced hyperglycemic mice. Thus, catalpol attenuates depressive-like behavior in pathological hyperglycemic state, and the antidepressant mechanism could at least be partly attributed to the upregulation of the PI3K/AKT/Nrf2/HO-1 signaling pathway in both brain regions, thus restoring the balance between oxidative and antioxidant damage. These data expanded the scientific understanding of catalpol and provided preclinical experimental evidence for its application.
Collapse
|
30
|
Rahman HS. Preclinical Drug Discovery in Colorectal Cancer: A Focus on Natural Compounds. Curr Drug Targets 2021; 22:977-997. [PMID: 33820517 DOI: 10.2174/1389450122666210405105206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/15/2021] [Accepted: 02/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is considered one of the most predominant and deadly cancer globally. Nowadays, the main clinical management for this cancer includes chemotherapy and surgery; however, these treatments result in the occurrence of drug resistance and severe side effects, and thus it is a crucial requirement to discover an alternative and potential therapy for CRC treatment. Numerous therapeutic cancers were initially recognized from natural metabolites utilized in traditional medicine, and several recent types of research have shown that many natural products own potential effects against CRC and may assist the action of chemotherapy for the treatment of CRC. It has been indicated that most patients are well tolerated by natural compounds without showing any toxicity signs even at high doses. Conventional chemotherapeutics interaction with natural medicinal compounds presents a new feature in cancer exploration and treatment. Most of the natural compounds overwhelm malignant cell propagation by apoptosis initiation of CRC cells and arresting of the cell cycle (especially at G, S, and G2/M phase) that result in inhibition of tumor growth. OBJECTIVE This mini-review aimed to focus on natural compounds (alkaloids, flavonoids, polysaccharides, polyphenols, terpenoids, lactones, quinones, etc.) that were identified to have anti- CRC activity in vitro on CRC cell lines and/or in vivo experiments on animal models. CONCLUSION Most of the studied active natural compounds possess anti-CRC activity via different mechanisms and pathways in vitro and in vivo that might be used as assistance by clinicians to support chemotherapy therapeutic strategy and treatment doses for cancer patients.
Collapse
Affiliation(s)
- Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, 46001 Sulaymaniyah, Iraq.,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaisee, Sulaimaniyah, Iraq
| |
Collapse
|
31
|
Catalpol exerts antiallergic effects in IgE/ovalbumin-activated mast cells and a murine model of ovalbumin-induced allergic asthma. Int Immunopharmacol 2021; 96:107782. [PMID: 34022666 DOI: 10.1016/j.intimp.2021.107782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/17/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022]
Abstract
Immunoglobulin E (IgE) and mast cells play important roles in the pathogenesis of allergic asthma. Catalpol, an iridoid glycoside, exerts many biological functions including anti-inflammatory activities. Herein, we investigated catalpol to determine both its antiallergic effects on IgE/ovalbumin (OVA)-stimulated mouse bone marrow-derived mast cells and its therapeutic actions in murine allergic asthma. We found that catalpol dramatically suppressed IgE/OVA-induced mast cell degranulation. Meanwhile, 5 ~ 100 μM of catalpol neither affected the expression level of the high-affinity receptor of IgE (FcεRI) by mast cells nor induced mast cell apoptosis. In addition, mRNA expression levels of inflammatory enzymes including cyclooxygenase (COX)-1, COX-2, and 5-lipoxygenase were downregulated. Administration of catalpol also suppressed production of prostaglandin D2 (PGD2), interleukin (IL)-6, and IL-13, while not affecting tumor necrosis factor (TNF)-α production. Further, catalpol pretreatment significantly attenuated the FcεRI-mediated Akt signaling pathway. In mice with IgE/OVA-induced asthma, oral administration of catalpol remarkably suppressed the production of OVA-specific IgE, the development of airway hyperresponsiveness (AHR), and the infiltration of eosinophils and neutrophils into the lungs. Histological studies demonstrated that catalpol substantially inhibited the recruitment of mast cells and increased mucus production in lung tissues. Catalpol-treated mice had significantly lower levels of helper T cell type 2 (Th2) cytokines (IL-4, IL-5, and IL-13), PGD2, eotaxin-1, and C-X-C chemokine ligand-1 (CXCL1) in bronchoalveolar lavage fluid (BALF) than did the allergic group. Collectively, these results indicated that the suppressive effects of catalpol on degranulation and mediator generation by mast cells were beneficial in treating allergic asthma.
Collapse
|
32
|
Shu Y, Liu X, Huang H, Wen Q, Shu J. Research progress of natural compounds in anti-liver fibrosis by affecting autophagy of hepatic stellate cells. Mol Biol Rep 2021; 48:1915-1924. [PMID: 33609264 PMCID: PMC7925445 DOI: 10.1007/s11033-021-06171-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Chronic liver diseases caused by various pathogenesis are marked by inflammatory infiltration and wound healing reaction, while their normal regeneration ability is impaired. The unbalance between the generation and the degradation of extracellular matrix (ECM) leads to collagen accumulation and develops into liver fibrosis. Inflammation, oxidative stress, and autophagy interact closely in the pathogenesis of hepatic fibrosis. Reactive Oxygen Species (ROS) can not only stimulate Kupffer cells to release massive inflammatory factors, but induce autophagy. However, the latter may suppress inflammatory reaction by inhibiting proinflammatory complex formation directly, and removing damaged organelles or pathogenic microorganism indirectly. At present, effective anti-fibrosis drugs are still lacking. Previous studies have found various natural compounds enabled liver protection through anti-inflammatory, antioxidant, and other mechanisms. In recent years, autophagy, a vital life activity, has been found to be involved in the mechanism of liver fibrosis. As a new target, developing anti-liver fibrosis drugs that regulate the activity of autophagy is very promising. In this review, we summarize the latest studies about natural compounds in the treatment of liver fibrosis by regulating autophagy.
Collapse
Affiliation(s)
- Yongxiang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Xuyou Liu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Haifeng Huang
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Qi Wen
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Jianchang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| |
Collapse
|
33
|
Yang F, Hou ZF, Zhu HY, Chen XX, Li WY, Cao RS, Li YX, Chen R, Zhang W. Catalpol Protects Against Pulmonary Fibrosis Through Inhibiting TGF-β1/Smad3 and Wnt/β-Catenin Signaling Pathways. Front Pharmacol 2021; 11:594139. [PMID: 33584272 PMCID: PMC7878558 DOI: 10.3389/fphar.2020.594139] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/29/2020] [Indexed: 12/30/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by fibroblast proliferation and extracellular matrix remodeling; however, the molecular mechanisms underlying its occurrence and development are not yet fully understood. Despite it having a variety of beneficial pharmacological activities, the effects of catalpol (CAT), which is extracted from Rehmannia glutinosa, in IPF are not known. In this study, the differentially expressed genes, proteins, and pathways of IPF in the Gene Expression Omnibus database were analyzed, and CAT was molecularly docked with the corresponding key proteins to screen its pharmacological targets, which were then verified using an animal model. The results show that collagen metabolism imbalance, inflammatory response, and epithelial-mesenchymal transition (EMT) are the core processes in IPF, and the TGF-β1/Smad3 and Wnt/β-catenin pathways are the key signaling pathways for the development of pulmonary fibrosis. Our results also suggest that CAT binds to TGF-βR1, Smad3, Wnt3a, and GSK-3β through hydrogen bonds, van der Waals bonds, and other interactions to downregulate the expression and phosphorylation of Smad3, Wnt3a, GSK-3β, and β-catenin, inhibit the expression of cytokines, and reduce the degree of oxidative stress in lung tissue. Furthermore, CAT can inhibit the EMT process and collagen remodeling by downregulating fibrotic biomarkers and promoting the expression of epithelial cadherin. This study elucidates several key processes and signaling pathways involved in the development of IPF, and suggests the potential value of CAT in the treatment of IPF.
Collapse
Affiliation(s)
- Fan Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen-Feng Hou
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hao-Yue Zhu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Xuan Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wan-Yang Li
- School of Public Health, Xiangya Medical College, Central South University, Changsha, China
| | - Ren-Shuang Cao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu-Xuan Li
- Second School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ru Chen
- Biomedical Research Institute of Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
34
|
Bi F, Xu Y, Chen G, Wang P. Anti-inflammatory and Anti-endoplasmic reticulum stress Effects of catalpol Against myocardial ischemia-reperfusion injury in streptozotocin-induced diabetic rats. AN ACAD BRAS CIENC 2020; 92:e20191148. [PMID: 33237136 DOI: 10.1590/0001-3765202020191148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/14/2020] [Indexed: 01/19/2023] Open
Abstract
The current study was designed to investigate the effects and the mechanism of catalpol on myocardial ischemia-reperfusion (MI/R) injury in a diabetic rat model. Male Sprague-Dawley rats were divided into DM + sham, DM +I/R, and DM +I/R + C groups and diabetes was induced using single injections of streptozotocin (STZ; 70 mg/kg; i.p). After confirming the induction of diabetes, rats were administered physiological saline and catalpol (10 mg/kg; i.p.) daily for 28 days. Subsequently, rats were subjected to left anterior descending (LAD) coronary artery occlusion for 30 min followed by reperfusion for 2 h. Haemodynamic parameters were recorded throughout surgery, and following sacrifice, hearts were isolated for biochemical, histopathological, and molecular analyses. Catalpol treatment significantly ameliorated MI/R injury by improving cardiac function, normalizing myocardial enzyme activities and markers of oxidative stress, and by maintaining myocardial architecture. Furthermore, expression levels of the inflammatory cytokines TNF-α and IL-6 were decreased in biochemical and immunohistochemical studies. Additionally, the cardioprotective effects of catalpol were partly related to reductions in myocardial endoplasmic reticulum stress (ERS). In conclusion, catalpol exerts cardioprotective effects in diabetic rats by attenuating inflammation and inhibiting ERS.
Collapse
Affiliation(s)
- Fangjie Bi
- Heart Center, Zibo Central Hospital, 255400 Shandong, China
| | - Yujia Xu
- Department of Echocardiography, Zibo Central Hospital, 255400 Shandong, China
| | - Guangxin Chen
- Department of Emergency, Zibo Central Hospital, 255400 Shandong, China
| | - Pan Wang
- Department of Pain Treatment, Zibo Central Hospital, 255400 Shandong, China
| |
Collapse
|
35
|
Effects of Catalpa Alcohol From Rehmannia glutinosa on Calcium-Binding Protein, Interleukin-1β, and Galectin-3 in Synovial Tissues of Rats With Knee Osteoarthritis. Int Surg 2020. [DOI: 10.9738/intsurg-d-20-00016.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objectives
We aimed to evaluate the effects of catalpa alcohol from Rehmannia glutinosa on the expressions of calcium-binding protein (S100A12), interleukin-1β (IL-1β), and galectin-3 in the synovium of rats with early knee osteoarthritis (KOA).
Methods
Fifty-two adult male Wistar rats aged 3 to 8 weeks were divided into normal control (n = 16), model (n = 12), low-dose (n = 12), and high-dose groups (n = 12). On the 10th day after modeling, 6 rats in normal control group and 6 in the other 3 groups were randomly selected. X-ray and 3-dimensional computed tomography (3D CT) images of the left knee joint were taken under live anesthesia. The joint cavity of sacrificed rats was opened to observe cartilage surface. After 28 consecutive days of administration, the synovial tissue of left knee joint was collected.
Results
The S100A12, IL-1β, and galectin-3 levels in synovial tissue were detected by immunohistochemistry and ELISA. There were articular cartilage defects in left knees. Radiologic examination showed significant joint space narrowing and hyperplasia, and 3D CT joint space value decreased (P < 0.05). The Mankins and OARSI scores of synovial histopathology were significantly different (P < 0.05). The S100A12, IL-1β, and galectin-3 levels in synovial tissue of the model group significantly exceeded those of the normal control group (P < 0.01). Compared with the model group, such levels of low-dose (P < 0.05) and high-dose groups (P < 0.01) were significantly lower.
Conclusions
The S100A12, IL-1β and galectin-3 levels in synovium tissue decreased with rising concentration of catalpa alcohol from R. glutinosa. Therefore, this drug is potentially suitable for inhibiting an inflammatory response to delay the progression of KOA.
Collapse
|
36
|
Wu L, Li H, Chen S, Wu X, Chen X, Wang F. Catalpol inhibits the proliferation, migration and metastasis of HCC cells by regulating miR‑140‑5p expression. Mol Med Rep 2020; 23:29. [PMID: 33179108 PMCID: PMC7673346 DOI: 10.3892/mmr.2020.11667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/15/2020] [Indexed: 01/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a frequent malignant tumor. Catalpol is a Chinese medicine extract with a number of pharmacologically active properties. The present study aimed to investigate the effects and mechanisms of catalpol in HCC. HCC cells were treated with catalpol in the presence or absence of microRNA (miR)-140-5p inhibitor, and assays to determine cell viability, proliferation, invasion and migration were performed. Reverse transcription-quantitative PCR and western blotting were performed to determine the mRNA and protein expression levels of miR-140-5p, vimentin, N-Cadherin and E-Cadherin. Moreover, cells were treated with catalpol in the absence or presence of transforming growth factor (TGF)-β1, and the cell morphology was observed under a microscope. The results demonstrated that catalpol inhibited cell proliferation, invasion and migration, and decreased the expression levels of vimentin and N-cadherin, but increased the expression levels of E-cadherin and miR-140-5p. Catalpol inhibited morphological changes in epithelial-mesenchymal transformation (EMT) of cells induced by TGF-β1. Following inhibition of miR-140-5p expression, the proliferation, invasion and migration of HCC cells were promoted, E-cadherin expression was decreased, and the levels of vimentin and N-cadherin were increased. The miR-140-5p inhibitor effectively reversed the inhibitory effect of catalpol on cell proliferation, invasion and migration. Thus, the results suggested that the antitumor potential of catalpol in HCC may be exerted by regulating the expression of miR-140-5p to inhibit proliferation, invasion, migration and EMT of HCC cells.
Collapse
Affiliation(s)
- Linsheng Wu
- Department of Geriatric Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - Haoxia Li
- Department of Geriatric Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - Shengyou Chen
- Department of Geriatric Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - Xiaoqiang Wu
- Department of Geriatric Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - Xiaomin Chen
- Department of Geriatric Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - Fangping Wang
- Department of Hepatobiliary Surgery, The People's Hospital of Xinchang, Shaoxing, Zhejiang 312500, P.R. China
| |
Collapse
|
37
|
Jiang X, Cao G, Gao G, Wang W, Zhao J, Gao C. Triptolide decreases tumor-associated macrophages infiltration and M2 polarization to remodel colon cancer immune microenvironment via inhibiting tumor-derived CXCL12. J Cell Physiol 2020; 236:193-204. [PMID: 32495392 DOI: 10.1002/jcp.29833] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/05/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Colon cancer is a common and deadly human digestive tract malignant tumor with poor prognosis. Immunotherapy has elicited tremendous success as a treatment modality for multiple solid tumors. Triptolide is extracted from the traditional Chinese medicine Tripterygium wilfordii Hook. F which shows various pharmacological actions including antitumor, anti-inflammatory, antimicrobial, antifibrosis, and antirheumatic. However, the influence of triptolide treatment on remodeling tumor immune microenvironment is still unknown in colon cancer. This study was aimed to investigate the therapeutic effect of triptolide treatment on colon cancer and the impact on tumor immune microenvironment and its underlying mechanism. We used CT26 subcutaneous tumors to conduct in vivo experiments and HT29, CT16, and Raw264.7 cells to perform in vitro assays. Triptolide had a therapeutic effect against colon cancer in vivo. Triptolide treatment distinctly inhibited the proliferation of colon cancer cells and induced apoptosis in vitro. In colon cancer immune microenvironment, triptolide treatment decreased the infiltration of tumor-associated macrophages through downregulating tumor-derived CXCL12 expression via nuclear factor kappa B and extracellular signal-regulated protein kinases 1 and 2 axis to remodel the immune microenvironment. Triptolide-educated colon cancers retarded the macrophages polarize to anti-inflammatory M2 status by decreasing the expression of Arg-1, CD206, and interleukin-10. Moreover, triptolide inhibited the migration of colon cancer cells via decreasing vascular endothelial growth factor expression. Our results identified the role of triptolide treatment in remodeling colon cancer immune microenvironment along with the distinct cytotoxicity function against colon cancer cells, which may provide the evidence for triptolide treatment in clinical.
Collapse
Affiliation(s)
- Xuan Jiang
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| | - Gang Cao
- Department of Respiratory Medicine, Hongze District People's Hospital, Hongze, Jiangsu, China
| | - Guangyi Gao
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| | - Wei Wang
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| | - Jiasheng Zhao
- Department of Oncology, Huaiyin Hospital of Huai'an City, Huai'an, China
| | - Chao Gao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
38
|
Qiao PF, Yao L, Zeng ZL. Catalpol‑mediated microRNA‑34a suppresses autophagy and malignancy by regulating SIRT1 in colorectal cancer. Oncol Rep 2020; 43:1053-1066. [PMID: 32323786 PMCID: PMC7057773 DOI: 10.3892/or.2020.7494] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common digestive tract tumors worldwide. Catalpol exerts inhibitory effects on the progression of several cancer types by regulating microRNAs (miRs). However, the precise role and carcinostatic mechanism of catalpol on CRC cells are poorly understood which limits the application of catalpol treatment. In the present study, miR-34a and sirtuin 1 (SIRT1) expression levels were detected in CRC tissues and CRC cell lines by RT-qPCR. Computational software analysis, luciferase assays and western blotting were used to demonstrate the downstream target of miR-34a in CRC cells. Effects of catalpol on cell viability, apoptosis, autophagic flux and the miR-34a/SIRT1 axis in the CRC cells were assessed by CCK-8 assay, flow cytometry, electron microscopy and western blotting, respectively. Whether the miR-34a/SIRT1 axis participated in catalpol-mediated autophagy and apoptosis was investigated. The effects of catalpol on the miR-34a/SIRT1 axis and malignant behavior were evaluated in a rat model of azoxymethane (AOM)-induced CRC. It was revealed that miR-34a expression levels were significantly decreased while SIRT1 was overexpressed in most of the CRC tissues and all the CRC cell lines. Clinically, a low level of miR-34a was correlated with poor clinicopathological characteristics in CRC patients. Catalpol reduced cell viability, suppressed autophagy, promoted apoptosis, and regulated the expression of SIRT1 by inducing miR-34a in vitro and in vivo. The autophagy-inhibiting effect of catalpol may be a mechanism to promote apoptosis of CRC cells. miR-34a mimic transfection resulted in autophagy-suppressive activity similar to that of catalpol, while the miR-34a inhibitor attenuated the antiautophagic effects of catalpol. In conclusion, miR-34a is involved in regulating catalpol-mediated autophagy and malignant behavior by directly inhibiting SIRT1 in CRC.
Collapse
Affiliation(s)
- Peng-Fei Qiao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Lei Yao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Zhao-Lin Zeng
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
39
|
Wu YS, Shi L, Liu XG, Li W, Wang R, Huang S, Li Y, Yan DL, Wang HY, Tian Y, Chen YM, Yang H. Chemical profiling of Callicarpa nudiflora and its effective compounds identification by compound-target network analysis. J Pharm Biomed Anal 2020; 182:113110. [PMID: 32004768 DOI: 10.1016/j.jpba.2020.113110] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 12/13/2022]
Abstract
Callicarpa nudiflora, belonging to the family Verbenaceae, is widely used to treat inflammation caused by bacterial infection.However, the underlying active substances of C. nudiflora against inflammation remains obscure. In this work, an ultra high-performance liquid chromatography (UHPLC) coupled with quadrupole time-of-flight mass spectrometry method was developed to characterize the ingredients in C. nudiflora, and a validated UHPLC coupled with triple quadrupole tandem mass spectrometry method was applied to quantify major components. As a result, a total of 96 chemical compounds were identified in C. nudiflora, and 26 compounds of them were further quantified in 34 batches of C. nudiflora. Based on the identified components from C. nudiflora, a compound-target network for the anti-inflammation effect was constructed by reverse docking target prediction, disease associated genes screening in DisGeNET and the protein-protein interaction from STRING. The compound-target network showed that C. nudiflora might exert anti-inflammation effect on the target of complement 3 and 5 in the pathway of cells and molecules involved in local acute inflammatory response, and 16 effective candidate compounds were found such as catalpol, acteoside, rutin, etc. This study provided an opportunity to deepen the understanding of the chemical composition and the potential anti-inflammatory mechanism of C. nudiflora.
Collapse
Affiliation(s)
- Yong-Sheng Wu
- Jiuzhitang Co., Ltd., No.339 West Tongzipo Road, High-Tech Industrial Development Zone, Changsha 410008, China
| | - Liu Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China
| | - Xin-Guang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China
| | - Wei Li
- Hainan Jiuzhitang Pharmaceutical Co., Ltd., A-12, Haikou national high-tech industrial development zone, Haikou 570311, China
| | - Rui Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China
| | - Sheng Huang
- Jiuzhitang Co., Ltd., No.339 West Tongzipo Road, High-Tech Industrial Development Zone, Changsha 410008, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China
| | - Dong-Lan Yan
- Jiuzhitang Co., Ltd., No.339 West Tongzipo Road, High-Tech Industrial Development Zone, Changsha 410008, China
| | - Hui-Ying Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China
| | - Yuan Tian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China
| | - Yan-Ming Chen
- Jiuzhitang Co., Ltd., No.339 West Tongzipo Road, High-Tech Industrial Development Zone, Changsha 410008, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24, Tongjia Lane, Nanjing 210009, China.
| |
Collapse
|
40
|
Bhattamisra SK, Yap KH, Rao V, Choudhury H. Multiple Biological Effects of an Iridoid Glucoside, Catalpol and Its Underlying Molecular Mechanisms. Biomolecules 2019; 10:E32. [PMID: 31878316 PMCID: PMC7023090 DOI: 10.3390/biom10010032] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Catalpol, an iridoid glucoside, is widely distributed in many plant families and is primarily obtained from the root of Rehmanniaglutinosa Libosch. Rehmanniaglutinosa is a plant very commonly used in Chinese and Korean traditional medicine for various disorders, including diabetes mellitus, neuronal disorders, and inflammation. Catalpol has been studied extensively for its biological properties both in vitro and in vivo. This review aims to appraise the biological effects of catalpol and their underlying mechanisms. An extensive literature search was conducted using the keyword "Catalpol" in the public domains of Google scholar, PubMed, and Scifinder. Catalpol exhibits anti-diabetic, cardiovascular protective, neuroprotective, anticancer, hepatoprotective, anti-inflammatory, and anti-oxidant effects in experimental studies. Anti-inflammatory and antioxidant properties are mostly related for its biological effect. However, some specific mechanisms are also elucidated. Elevated serotonin and BDNF level by catalpol significantly protect against depression and neurodegeneration. Catalpol demonstrated an increased mitochondrial biogenesis and activation of PI3K/Akt pathway for insulin sensitizing effect. Further, its cardiovascular protective effect was linked to PI3K/Akt, apelin/APJ and Jak-Stat pathway. Catalpol produced a significant reduction in cell proliferation and an increase in apoptosis in different cancer conditions. Overall, catalpol demonstrated multiple biological effects due to its numerous mechanisms including anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Kah Heng Yap
- School of Post graduate studies, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia; (K.H.Y.); (V.R.)
| | - Vikram Rao
- School of Post graduate studies, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia; (K.H.Y.); (V.R.)
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia;
| |
Collapse
|
41
|
Aiello P, Sharghi M, Mansourkhani SM, Ardekan AP, Jouybari L, Daraei N, Peiro K, Mohamadian S, Rezaei M, Heidari M, Peluso I, Ghorat F, Bishayee A, Kooti W. Medicinal Plants in the Prevention and Treatment of Colon Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2075614. [PMID: 32377288 PMCID: PMC7187726 DOI: 10.1155/2019/2075614] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/03/2019] [Indexed: 01/04/2023]
Abstract
The standard treatment for cancer is generally based on using cytotoxic drugs, radiotherapy, chemotherapy, and surgery. However, the use of traditional treatments has received attention in recent years. The aim of the present work was to provide an overview of medicinal plants effective on colon cancer with special emphasis on bioactive components and underlying mechanisms of action. Various literature databases, including Web of Science, PubMed, and Scopus, were used and English language articles were considered. Based on literature search, 172 experimental studies and 71 clinical cases on 190 plants were included. The results indicate that grape, soybean, green tea, garlic, olive, and pomegranate are the most effective plants against colon cancer. In these studies, fruits, seeds, leaves, and plant roots were used for in vitro and in vivo models. Various anticolon cancer mechanisms of these medicinal plants include induction of superoxide dismutase, reduction of DNA oxidation, induction of apoptosis by inducing a cell cycle arrest in S phase, reducing the expression of PI3K, P-Akt protein, and MMP as well; reduction of antiapoptotic Bcl-2 and Bcl-xL proteins, and decrease of proliferating cell nuclear antigen (PCNA), cyclin A, cyclin D1, cyclin B1 and cyclin E. Plant compounds also increase both the expression of the cell cycle inhibitors p53, p21, and p27, and the BAD, Bax, caspase 3, caspase 7, caspase 8, and caspase 9 proteins levels. In fact, purification of herbal compounds and demonstration of their efficacy in appropriate in vivo models, as well as clinical studies, may lead to alternative and effective ways of controlling and treating colon cancer.
Collapse
Affiliation(s)
- Paola Aiello
- Council for Agricultural Research and Economics, Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy
- Department of Physiology and Pharmacology “V. Erspamer”, La Sapienza University of Rome, Rome, Italy
| | - Maedeh Sharghi
- Nursing and Midwifery School, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Azam Pourabbasi Ardekan
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Leila Jouybari
- Nursing Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Nahid Daraei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Khadijeh Peiro
- Department of Biology, Faculty of Sciences, Shahid Chamran University, Ahvaz, Iran
| | - Sima Mohamadian
- Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdiyeh Rezaei
- Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Heidari
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ilaria Peluso
- Council for Agricultural Research and Economics, Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy
| | - Fereshteh Ghorat
- Traditional and Complementary Medicine Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL 34211, USA
| | - Wesam Kooti
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
42
|
Zhao L, Wang Y, Liu Q. Catalpol inhibits cell proliferation, invasion and migration through regulating miR-22-3p/MTA3 signalling in hepatocellular carcinoma. Exp Mol Pathol 2019; 109:51-60. [DOI: 10.1016/j.yexmp.2019.104265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/13/2019] [Accepted: 05/24/2019] [Indexed: 01/19/2023]
|
43
|
Hong M, Shi H, Wang N, Tan HY, Wang Q, Feng Y. Dual Effects of Chinese Herbal Medicines on Angiogenesis in Cancer and Ischemic Stroke Treatments: Role of HIF-1 Network. Front Pharmacol 2019; 10:696. [PMID: 31297056 PMCID: PMC6606950 DOI: 10.3389/fphar.2019.00696] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1)–induced angiogenesis has been involved in numerous pathological conditions, and it may be harmful or beneficial depending on the types of diseases. Exploration on angiogenesis has sparked hopes in providing novel therapeutic approaches on multiple diseases with high mortality rates, such as cancer and ischemic stroke. The HIF-1 pathway is considered to be a major regulator of angiogenesis. HIF-1 seems to be involved in the vascular formation process by synergistic correlations with other proangiogenic factors in cancer and cerebrovascular disease. The regulation of HIF-1–dependent angiogenesis is related to the modulation of HIF-1 bioactivity by regulating HIF-1α transcription or protein translation, HIF-1α DNA binding, HIF-1α and HIF-1α dimerization, and HIF-1 degradation. Traditional Chinese herbal medicines have a long history of clinical use in both cancer and stroke treatments in Asia. Growing evidence has demonstrated potential proangiogenic benefits of Chinese herbal medicines in ischemic stroke, whereas tumor angiogenesis could be inhibited by the active components in Chinese herbal medicines. The objective of this review is to provide comprehensive insight on the effects of Chinese herbal medicines on angiogenesis by regulating HIF-1 pathways in both cancer and ischemic stroke.
Collapse
Affiliation(s)
- Ming Hong
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglian Shi
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS, United States
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Hor-Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
44
|
Awad D, Prattes M, Kofler L, Rössler I, Loibl M, Pertl M, Zisser G, Wolinski H, Pertschy B, Bergler H. Inhibiting eukaryotic ribosome biogenesis. BMC Biol 2019; 17:46. [PMID: 31182083 PMCID: PMC6558755 DOI: 10.1186/s12915-019-0664-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ribosome biogenesis is a central process in every growing cell. In eukaryotes, it requires more than 250 non-ribosomal assembly factors, most of which are essential. Despite this large repertoire of potential targets, only very few chemical inhibitors of ribosome biogenesis are known so far. Such inhibitors are valuable tools to study this highly dynamic process and elucidate mechanistic details of individual maturation steps. Moreover, ribosome biogenesis is of particular importance for fast proliferating cells, suggesting its inhibition could be a valid strategy for treatment of tumors or infections. RESULTS We systematically screened ~ 1000 substances for inhibitory effects on ribosome biogenesis using a microscopy-based screen scoring ribosomal subunit export defects. We identified 128 compounds inhibiting maturation of either the small or the large ribosomal subunit or both. Northern blot analysis demonstrates that these inhibitors cause a broad spectrum of different rRNA processing defects. CONCLUSIONS Our findings show that the individual inhibitors affect a wide range of different maturation steps within the ribosome biogenesis pathway. Our results provide for the first time a comprehensive set of inhibitors to study ribosome biogenesis by chemical inhibition of individual maturation steps and establish the process as promising druggable pathway for chemical intervention.
Collapse
Affiliation(s)
- Dominik Awad
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
- Present address: Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Prattes
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Lisa Kofler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Ingrid Rössler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Mathias Loibl
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Melanie Pertl
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Gertrude Zisser
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Brigitte Pertschy
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria.
| | - Helmut Bergler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria.
| |
Collapse
|
45
|
Parveen A, Subedi L, Kim HW, Khan Z, Zahra Z, Farooqi MQ, Kim SY. Phytochemicals Targeting VEGF and VEGF-Related Multifactors as Anticancer Therapy. J Clin Med 2019; 8:E350. [PMID: 30871059 PMCID: PMC6462934 DOI: 10.3390/jcm8030350] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 02/06/2023] Open
Abstract
The role of vascular endothelial growth factor (VEGF) in cancer cells is not limited to angiogenesis; there are also multiple factors, such as neuropilins (non-tyrosine kinases receptors), tyrosine kinases receptors, immunodeficiencies, and integrins, that interact with VEGF signaling and cause cancer initiation. By combating these factors, tumor progression can be inhibited or limited. Natural products are sources of several bioactive phytochemicals that can interact with VEGF-promoting factors and inhibit them through various signaling pathways, thereby inhibiting cancer growth. This review provides a deeper understanding of the relation and interaction of VEGF with cancer-promoting factors and phytochemicals in order to develop multi-targeted cancer prevention and treatment.
Collapse
Affiliation(s)
- Amna Parveen
- Department of Pharmacognosy, Faculty of Pharmaceutical Science, Government College University, Faisalabad, Faisalabad 38000, Pakistan.
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Lalita Subedi
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Heung Wan Kim
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Zahra Khan
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Zahra Zahra
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology, Sector H-12, Islamabad 44000, Pakistan.
| | | | - Sun Yeou Kim
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
- Gachon Institute of Pharmaceutical Science, Gachon University, No. 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea.
| |
Collapse
|
46
|
Investigation of dynamic accumulation and regularity of nine glycosides and saccharides in
Rehmannia glutinosa
by rapid quantitative analysis technology. J Sep Sci 2019; 42:1489-1499. [DOI: 10.1002/jssc.201801185] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/28/2022]
|
47
|
Wang Z, Lu Y, Sheng B, Ding Y, Cheng X. Catalpol inhibits TGF-β1-induced epithelial-mesenchymal transition in human non-small-cell lung cancer cells through the inactivation of Smad2/3 and NF-κB signaling pathways. J Cell Biochem 2019; 120:2251-2258. [PMID: 30203551 DOI: 10.1002/jcb.27535] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/01/2018] [Indexed: 01/24/2023]
Abstract
Catalpol, one of the main active ingredients isolated from Rehmannia glutinosa, was reported to possess anticancer activity. However, the role of catalpol in transforming growth factor β1 (TGF-β1)-induced epithelial-mesenchymal transition (EMT) in human non-small-cell lung cancer (NSCLC) cells has not been elucidated. The objective of this study was to investigate the effect of catalpol on EMT in human NSCLC cells. Our results showed that catalpol significantly inhibited the TGF-β1-induced cell migration and invasion of A549 cells, as well as repressed matrix metalloproteinase (MMP)2 and MMP9 expression induced by TGF-β1 in A549 cells. In addition, catalpol markedly repressed the EMT process in A549 cells in response to TGF-β1. Furthermore, catalpol prevented the activation of Smad2/3 and nuclear factor κB (NF-κB) signaling pathways induced by TGF-β1 in A549 cells. In conclusion, these findings indicated that catalpol inhibits TGF-β1-induced EMT in human NSCLC cells through the inactivation of Smad2/3 and NF-κB signaling pathways. Thus, catalpol may be a promising agent for the treatment of NSCLC.
Collapse
Affiliation(s)
- Zuopei Wang
- Department of Thoracic Surgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Yi Lu
- Department of Thoracic Surgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Bo Sheng
- Department of Thoracic Surgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Yi Ding
- Department of Thoracic Surgery, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Xiaoke Cheng
- Department of Scientific Research, Shanghai Pudong New Area People's Hospital, Shanghai, China
| |
Collapse
|
48
|
Sosorburam D, Wu ZG, Zhang SC, Hu P, Zhang HY, Jiang T, Ahiasi-Mensah J, He X. Therapeutic effects of traditional Chinese herbal prescriptions for primary dysmenorrhea. CHINESE HERBAL MEDICINES 2019. [DOI: 10.1016/j.chmed.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
49
|
Shen X, Tao Y, Yang Y, Wang R, Fang J, Lian M. Combination of TPF regimen and cinobufotalin inhibits proliferation and induces apoptosis in human hypopharyngeal and laryngeal squamous cell carcinoma cells. Onco Targets Ther 2018; 12:341-348. [PMID: 30643431 PMCID: PMC6317469 DOI: 10.2147/ott.s185428] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Hypopharyngeal squamous cell carcinoma and laryngeal squamous cell carcinoma have a potential risk of metastasis and local recurrence. It is urgent to determine novel effective therapy. Methods UMSCC5 and FADU cells were treated with TPF regimen (a mixture comprising paclitaxel, cisplatin, and 5-fluorouracil), cinobufotalin, and combination. Cell counting kit-8 (CCK-8), clone formation assays, and flow cytometry were performed to investigate whether combination of cinobufotalin and TPF can exhibit synergistic effects in reducing tumor growth. Results CCK-8 assay and IC50 analysis showed that the TPF and cinobufotalin could suppress viability of UMSCC5 and FADU cells, implying the strong antitumor effect. The synergism between TPF and cinobufotalin was further verified by the CCK-8 and clone formation assays showing the TPF- and cinobufotalin-suppressed cell proliferation synergistically. Notably, flow cytometry showed that the combination also promoted apoptosis synergistically. Conclusion TPF regimen combining cinobufotalin suppressed proliferation and promoted apoptosis synergistically in hypopharyngeal squamous cell carcinoma and laryngeal squamous cell carcinoma cells.
Collapse
Affiliation(s)
- Xixi Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China, ;
| | - Yong Tao
- Department of Pharmacy, Liaocheng Third People's Hospital, Liaocheng, Shandong 252000, China
| | - Yifan Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China, ;
| | - Ru Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China, ;
| | - Jugao Fang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China, ;
| | - Meng Lian
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China, ;
| |
Collapse
|
50
|
Liang J, Wu J, Wang F, Zhang P, Zhang X. Semaphoring 4D is required for the induction of antioxidant stress and anti-inflammatory effects of dihydromyricetin in colon cancer. Int Immunopharmacol 2018; 67:220-230. [PMID: 30562683 DOI: 10.1016/j.intimp.2018.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022]
Abstract
Semaphorin 4D (Sema4D) has been involved in cancer progression, the expression of which is associated with the poor clinical outcomes of some cancer patients. Dihydromyricetin (DMY) has antitumor potentials for different types of human cancer cells. However, the pharmacological effects of DMY on colon cancer (CC) or the regulatory effects of Sema4D on this process remain largely unknown. In the present study, we aimed to evaluate the effects of DMY on CC, and to elucidate the role of Sema4D in DMY-induced antitumor effects. DMY inhibited the proliferation and growth of Colo-205 colon cancer cells significantly in vivo and in vitro. DMY inhibited reactive oxygen species (ROS) and malondialdehyde (MDA) levels, but increased glutathione (GSH) level. Moreover, the activities of antioxidant enzymes catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR) and heme oxygenase 1 (HO-1) were enhanced by DMY treatment in vitro, showing strong anti-oxidative stress effect. In addition, DMY inhibited the secretion of interleukin 1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8) and tumor necrosis factor (TNF-α) in the supernatant of Colo-205 culture medium. Besides, the expressions of cyclooxygenase (COX-2) and inducible nitric oxide synthase (iNOS) were suppressed by DMY in dose-dependent manners in vivo, showing potent anti-inflammatory effect. Further investigations showed that DMY suppressed the expression and secretion of Sema4D in Colo-205 cells and tissues. Interestingly, overexpression of Sema4D significantly weakened the regulatory effects of DMY on oxidative stress. Furthermore, overexpression of Sema4D significantly attenuated the anti-inflammatory effects of DMY. Collectively, we drew a conclusion that the anti-colon cancer effect of DMY was attributed to its negative modulation on oxidative stress and inflammation via suppression of Sema4D. The findings broaden the width and depth of molecular mechanisms involved in the DMY action, facilitating the development of DMY in anti-colon cancer therapies.
Collapse
Affiliation(s)
- Jun Liang
- Oncology Medicine Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Jing Wu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Fei Wang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Pengfei Zhang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xuemei Zhang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|