1
|
Baidya R, Sarkar B. A systematic review of the traditional uses, chemistry, and curative aptitude of echinacoside-a phenylethanoid glycoside. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2071-2106. [PMID: 39361172 DOI: 10.1007/s00210-024-03460-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/12/2024] [Indexed: 03/19/2025]
Abstract
Echinacoside (ECH), a naturally occurring water-soluble phenylethanoid glycoside, is one of the primary bioactive compounds present in several plant species, such as Echinacea, Cistanche, Plantago, Rosa, Buddleja, and Rehmannia. Research has revealed that these plants, rich in ECH, have diverse traditional uses and pharmacological activities, like anti-diabetic, anti-inflammatory, anti-fatigue, anti-allergic, anti-ageing, anti-skin glycation, analgesic, wound healing, and aphrodisiac properties. Among other activities, ophthalmic, haematopoiesis, pulmonary, anti-bacterial, anti-protozoal, anti-fungal, and anti-viral effects of ECH have been reported. Chemically, the compound comprises caffeic acid glycoside containing a trisaccharide that includes two glucose and one rhamnose unit. These units are linked through glycosidic bonds to a caffeic acid and a dihydroxyphenylethanol (hydroxytyrosol) residue, which are connected to the central rhamnose. The biosynthesis of ECH has been reported to start with forming L-phenylalanine and tyrosine precursors via the shikimic acid pathway. The structure-activity relationship of ECH has shown that various functional groups in the structure, particularly phenolic hydroxyl groups, are crucial for antioxidant activities. Similarly, in silico studies have revealed that ECH binds to different receptors, like Kelch-like ECH-associated protein 1 (Keap1), receptor for advanced glycation end products (RAGE), etc., to affect various pharmacological activities. The ECH contents in the reported plants often own these multifaceted properties, highlighting their importance in clinical research. Evident from its therapeutic efficacy, there is a huge potential for a comprehensive understanding of the mechanisms of actions of ECH, which underscores the need for more research in this area. Thus, this review is a compendium of the latest literature to analyse the existing knowledge on ECH, encompassing its distribution, traditional uses, extraction, chemical constituents, biosynthesis, pharmacological activities, structure-activity relationship, and in silico studies, following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.
Collapse
Affiliation(s)
- Ritika Baidya
- Group Polyphenol-BIT, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India
| | - Biswatrish Sarkar
- Group Polyphenol-BIT, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, India.
| |
Collapse
|
2
|
Wang X, Tan B, Liu J, Wang J, Chen M, Yang Q, Zhang X, Li F, Wei Y, Wu K, Ren G, Li H. Echinacoside inhibits tumor immune evasion by downregulating inducible PD-L1 and reshaping tumor immune landscape in breast and colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156188. [PMID: 39488102 DOI: 10.1016/j.phymed.2024.156188] [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: 06/12/2024] [Revised: 10/01/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Targeting PD-L1 has become a crucial approach in tumor immunotherapy. Echinacoside (ECH) is a natural compound known for its extensive biological activities, its impact on antitumor immunity remains uncertain. PURPOSE This work was designed to assess the effects of ECH on the PD-L1/PD-1-mediated tumor immune evasion and its underlying mechanisms. METHODS Flow cytometry and RT-qPCR were utilized to explore the influence of ECH on PD-L1 expression. Western blot was employed to examine the mechanism by which ECH might modulate PD-L1 expression. Flow cytometry was conducted to evaluate the influence of ECH therapy, or the synergistic effects of ECH combined with immune checkpoint blockade (ICB) on tumor immune microenvironment (TIME) in tumor-burden mice. Blood biochemistry tests were used to evaluate the safety of ECH treatment. RESULTS ECH downregulated both the protein and mRNA expression levels of IFN-γ-induced PD-L1 through JAK/STAT1/IRF1 signaling pathway. ECH treatment upregulated the infiltration of IFN-γ+CD8+ T cells and Ki-67+CD8+ T cells, lowered the frequency of TIM-3+PD-1+ T cells, promoted the infiltration of effector CD4+ T cells and total CD8+ T cells while suppressed the percentage of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC). Moreover, the combination of ECH and anti-PD-1 or anti-CTLA-4 therapy exhibited synergistic anti-tumor effects, reshaping TIME. Blood biochemistry tests unveiled that ECH did not show additional toxicity. CONCLUSION ECH upregulates the expression of inducible PD-L1 through the JAK/STAT1/IRF1 signaling pathway, enhances T cell function, and reshapes the tumor immune landscape into an anti-tumor phenotype. Importantly, ECH markedly enhances the efficacy of ICB treatment, indicating its potential application in anti-tumor therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Binxin Tan
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Oncology, The Affiliated Banan's Hospital of Chongqing Medical University, Chongqing 401320, China
| | - Jiazhou Liu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jing Wang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of the University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei 230000, China
| | - Mingjing Chen
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qian Yang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiang Zhang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fan Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuxian Wei
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ke Wu
- Department of Pharmacy, The Affiliated Dazu's Hospital of Chongqing Medical University, Chongqing 402360, China.
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
3
|
Fu Q, Yang J, Jiang H, Lin S, Qin H, Zhao J, Wang Y, Liu M. Effect of photobiomodulation on alleviating primary dysmenorrhea caused by PGF 2α. JOURNAL OF BIOPHOTONICS 2024; 17:e202300448. [PMID: 38348528 DOI: 10.1002/jbio.202300448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 05/04/2024]
Abstract
Photobiomodulation (PBM) has attracted widespread attention in suppressing various pain and inflammation. Primary dysmenorrhea (PD) primarily occurs in adolescents and adult females, and the limited effectiveness and side effects of conventional treatments have highlighted the urgent need to develop and identify new adjunct therapeutic strategies. In this work, the results of pain and PGs demonstrated that 850 nm, 630 nm, and 460 nm all exhibited pain inhibition, decreased PGF2α and upregulated PGE2, while 630 nm PBM has better effectiveness. Then to explore the underlying biological mechanisms of red light PBM on PD, we irradiated prostaglandin-F2α induced HUSM cells and found that low-level irradiance can restore intracellular calcium ion, ROS, ATP, and MMP levels to normal levels. And, red light enhanced cell viability and promoted cell proliferation for normal HUSM cells. Therefore, this study proposes that red light PBM may be a promising approach for the future clinical treatment of PD.
Collapse
Affiliation(s)
- Qiqi Fu
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Jiali Yang
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Hui Jiang
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Shangfei Lin
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Haokuan Qin
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Jie Zhao
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Yanqing Wang
- School of Basic Medical Science, Fudan University, Shanghai, China
| | - Muqing Liu
- School of Information Science and Technology, Fudan University, Shanghai, China
- Zhongshan Fudan Joint Innovation Center, Zhongshan, Guangdong Province, China
| |
Collapse
|
4
|
Ahmadi F, Kariman K, Mousavi M, Rengel Z. Echinacea: Bioactive Compounds and Agronomy. PLANTS (BASEL, SWITZERLAND) 2024; 13:1235. [PMID: 38732450 PMCID: PMC11085449 DOI: 10.3390/plants13091235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024]
Abstract
For centuries, medicinal plants have been used as sources of remedies and treatments for various disorders and diseases. Recently, there has been renewed interest in these plants due to their potential pharmaceutical properties, offering natural alternatives to synthetic drugs. Echinacea, among the world's most important medicinal plants, possesses immunological, antibacterial, antifungal, and antiviral properties. Nevertheless, there is a notable lack of thorough information regarding the echinacea species, underscoring the vital need for a comprehensive review paper to consolidate existing knowledge. The current review provides a thorough analysis of the existing knowledge on recent advances in understanding the physiology, secondary metabolites, agronomy, and ecology of echinacea plants, focusing on E. purpurea, E. angustifolia, and E. pallida. Pharmacologically advantageous effects of echinacea species on human health, particularly distinguished for its ability to safeguard the nervous system and combat cancer, are discussed. We also highlight challenges in echinacea research and provide insights into diverse approaches to boost the biosynthesis of secondary metabolites of interest in echinacea plants and optimize their large-scale farming. Various academic databases were employed to carry out an extensive literature review of publications from 2001 to 2024. The medicinal properties of echinacea plants are attributed to diverse classes of compounds, including caffeic acid derivatives (CADs), chicoric acid, echinacoside, chlorogenic acid, cynarine, phenolic and flavonoid compounds, polysaccharides, and alkylamides. Numerous critical issues have emerged, including the identification of active metabolites with limited bioavailability, the elucidation of specific molecular signaling pathways or targets linked to echinacoside effects, and the scarcity of robust clinical trials. This raises the overarching question of whether scientific inquiry can effectively contribute to harnessing the potential of natural compounds. A systematic review and analysis are essential to furnish insights and lay the groundwork for future research endeavors focused on the echinacea natural products.
Collapse
Affiliation(s)
- Fatemeh Ahmadi
- UWA School of Agriculture and Environment, The University of Western Australia, Perth, WA 6009, Australia; (K.K.); (M.M.); (Z.R.)
| | - Khalil Kariman
- UWA School of Agriculture and Environment, The University of Western Australia, Perth, WA 6009, Australia; (K.K.); (M.M.); (Z.R.)
| | - Milad Mousavi
- UWA School of Agriculture and Environment, The University of Western Australia, Perth, WA 6009, Australia; (K.K.); (M.M.); (Z.R.)
| | - Zed Rengel
- UWA School of Agriculture and Environment, The University of Western Australia, Perth, WA 6009, Australia; (K.K.); (M.M.); (Z.R.)
- Institute for Adriatic Crops and Karst Reclamation, 21000 Split, Croatia
| |
Collapse
|
5
|
Tang Y, Zhao F, Zhang X, Niu Y, Liu X, Bu R, Ma Y, Wu G, Li B, Yang H, Wu J. Cistanche phenylethanoid glycosides induce apoptosis and pyroptosis in T-cell lymphoma. Am J Cancer Res 2024; 14:1338-1352. [PMID: 38590417 PMCID: PMC10998756 DOI: 10.62347/gezw9659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Cistanche deserticola, known for its extensive history in Traditional Chinese Medicine (TCM), is valued for its therapeutic properties. Recent studies have identified its anticancer capabilities, yet the mechanisms underlying these properties remain to be fully elucidated. In this study, we determined that a mixture of four cistanche-derived phenylethanoid glycosides (CPhGs), echinacoside, acteoside, 2-acetylacteoside, and cistanoside A, which are among the main bioactive compounds in C. deserticola, eliminated T-cell lymphoma (TCL) cells by inducing apoptosis and pyroptosis in vitro and attenuated tumor growth in vivo in a xenograft mouse model. At the molecular level, these CPhGs elevated P53 by inhibiting the SIRT2-MDM2/P300 and PI3K/AKT carcinogenic axes and activating PTEN-Bax tumor-suppressing signaling. Moreover, CPhGs activated noncanonical and alternative pathways to trigger pyroptosis. Interestingly, CPhGs did not activate canonical NLRP3-caspase-1 pyroptotic signaling pathway; instead, CPhGs suppressed the inflammasome factor NLRP3 and the maturation of IL-1β. Treatment with a caspase-1/4 inhibitor and silencing of Gasdermin D (GSDMD) or Gasdermin E (GSDME) partially rescued CPhG-induced cell death. Conversely, forced expression of NLRP3 restored cell proliferation. In summary, our results indicate that CPhGs modulate multiple signaling pathways to achieve their anticancer properties and perform dual roles in pyroptosis and NLRP3-driven proliferation. This study offers experimental support for the potential application of CPhGs in the treatment of TCL.
Collapse
Affiliation(s)
- Ying Tang
- School of Life Sciences, Inner Mongolia UniversityHohhot, Inner Mongolia, China
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Fangxin Zhao
- School of Life Sciences, Inner Mongolia UniversityHohhot, Inner Mongolia, China
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Xuan Zhang
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Yan Niu
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Xiulan Liu
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Renqiqige Bu
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Yunlong Ma
- School of Life Sciences, Inner Mongolia Agricultural UniversityHohhot, Inner Mongolia, China
| | - Geyemuri Wu
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Beibei Li
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Hongxin Yang
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| | - Jianqiang Wu
- School of Life Sciences, Inner Mongolia UniversityHohhot, Inner Mongolia, China
- College of Basic Medicine, Inner Mongolia Medical UniversityHohhot, Inner Mongolia, China
| |
Collapse
|
6
|
Liang J, Chen T, Xu H, Wang T, Gong Q, Li T, Liu X, Wang J, Wang Y, Xiong L. Echinacoside Exerts Antihepatic Fibrosis Effects in High-Fat Mice Model by Modulating the ACVR2A-Smad Pathway. Mol Nutr Food Res 2024; 68:e2300553. [PMID: 38366962 DOI: 10.1002/mnfr.202300553] [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: 08/02/2023] [Revised: 01/14/2024] [Indexed: 02/19/2024]
Abstract
SCOPE Nonalcoholic steatohepatitis (NASH) is an increasingly common chronic liver disease in which hepatic fibrosis is the major pathological change. The transforming growth factor β (TGF-β)/mall mothers against decapentaplegic (Smad) signaling is the main effector of fibrosis. Although the antifibrotic effect of echinacoside (Ech) on the liver has been indicated previously, the cellular and molecular mechanisms remain unclear. This study aims to investigate both in vivo and in vitro antifibrotic properties of Ech. METHODS AND RESULTS Cell viability and scratch/wound assays show that Ech significantly inhibits the proliferation, migration, and activation of human hepatic stellate LX-2 cells. In mice with high-fat diet-induced hepatic fibrosis, Ech treatment attenuates the progression of liver injury, inflammation, and fibrosis. Furthermore, transcriptome analysis and subsequent functional validation demonstrate that Ech achieves antifibrotic effects by the activin receptor type-2A (ACVR2A)-mediated TGF-β1/Smad signaling pathway; ultimately, ACVR2A is demonstrated to be an important target for hepatic fibrosis by inhibiting and inducing the expression of ACVR2A in LX-2 cells. CONCLUSION Ech exerts potent antifibrotic effects by inhibiting the ACVR2A-mediated TGF-β1/Smad signaling axis and may serve as an alternative treatment for hepatic fibrosis.
Collapse
Affiliation(s)
- Jie Liang
- Department of Medicine, Linfen Vocational and Technical College, Linfen, Shanxi, 041000, China
| | - Ting Chen
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Honglei Xu
- Medical Security Center, The No. 983th Hospital of Joint Logistics Support Forces of Chinese PLA, Tianjin, 300142, China
| | - Tingfang Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Qi Gong
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Tingting Li
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiaoyan Liu
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jing Wang
- Department of Pharmacy, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Yun Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Liyan Xiong
- School of Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
7
|
Xin X, Cheng X, Zeng F, Xu Q, Hou L. The Role of TGF-β/SMAD Signaling in Hepatocellular Carcinoma: from Mechanism to Therapy and Prognosis. Int J Biol Sci 2024; 20:1436-1451. [PMID: 38385079 PMCID: PMC10878151 DOI: 10.7150/ijbs.89568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/15/2024] [Indexed: 02/23/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, with high incidence and mortality, accounting for approximately 90% of liver cancer. The development of HCC is a complex process involving the abnormal activation or inactivation of multiple signaling pathways. Transforming growth factor-β (TGF-β)/Small mothers against decapentaplegic (SMAD) signaling pathway regulates the development of HCC. TGF-β activates intracellular SMADs protein through membrane receptors, resulting in a series of biological cascades. Accumulating studies have demonstrated that TGF-β/SMAD signaling plays multiple regulatory functions in HCC. However, there is still controversy about the role of TGF-β/SMAD in HCC. Because it involves different pathogenic factors, disease stages, and cell microenvironment, as well as upstream and downstream relationships with other signaling pathways. This review will summary the regulatory mechanism of the TGF-β/SMAD signaling pathway in HCC, involving the regulation of different pathogenic factors, different disease stages, different cell populations, microenvironments, and the interaction with microRNAs. In addition, we also introduced small molecule inhibitors, therapeutic vaccines, and traditional Chinese medicine extracts based on targeting the TGF-β/SMAD signaling pathway, which will provide future research direction for HCC therapy targeting the TGF-β/SMAD signaling pathway.
Collapse
Affiliation(s)
- Xin Xin
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Xiyu Cheng
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan province, China
| | - Qing Xu
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Lingling Hou
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
8
|
Zhang B, Zhou B, Huang G, Huang J, Lin X, Li Z, Lian Y, Huang Q, Ye Y. Nitidine chloride inhibits G2/M phase by regulating the p53/14-3-3 Sigma/CDK1 axis for hepatocellular carcinoma treatment. Heliyon 2024; 10:e24012. [PMID: 38283241 PMCID: PMC10818205 DOI: 10.1016/j.heliyon.2024.e24012] [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: 10/22/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Background Liver cancer had become the sixth most common cancer. Nitidine chloride (NC) has demonstrated promising anti-HCC properties; however, further elucidation of its mechanism of action is necessary. Methods The anti-HCC targets of NC were identified through the utilization of multiple databases and ChIPs data analysis. The GO and KEGG analyses to determine the specific pathway affected by NC. The Huh 7 and Hep G2 cells were subjected to a 24-h treatment with NC, followed by evaluating the impact of NC on cell proliferation and cell cycle. The involvement of the p53/14-3-3 Sigma/CDK1 axis in HCC cells was confirmed by qPCR and WB analysis of the corresponding genes and proteins. Results The GO and KEGG analysis showed the targets were related to cell cycle and p53 signaling pathways. In vitro experiments showed that NC significantly inhibited the proliferation of HCC cells and induced G2/M phase arrest. In addition, qPCR and WB experiments showed that the expression of p53 in HCC cells increased after NC intervention, while the expression of 14-3-3 Sigma and CDK1 decreased. Conclusion NC can inhibit the proliferation of HCC cells and induce G2/M cell cycle arrest, potentially by regulating the p53/14-3-3 Sigma/CDK1 axis.
Collapse
Affiliation(s)
- Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Bo Zhou
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Guihong Huang
- Department of Pharmacy, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541199, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, 541199, China
| | - Jing'an Huang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Xiaoxin Lin
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Zonghuai Li
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Yuanchu Lian
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Qiujie Huang
- Guangxi University of Chinese Medicine, Teaching Experiment and Training Center, Nanning, China
| | - Yong Ye
- School of Pharmacy, Guangxi Medical University, Guangxi, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, China
| |
Collapse
|
9
|
Wei J, Zheng Z, Hou X, Jia F, Yuan Y, Yuan F, He F, Hu L, Zhao L. Echinacoside inhibits colorectal cancer metastasis via modulating the gut microbiota and suppressing the PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116866. [PMID: 37429503 DOI: 10.1016/j.jep.2023.116866] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/10/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Echinacoside (ECH) is the dominant phenylethanoid glycoside-structured compound identified from our developed herbal formula Huangci granule, which has been previously reported to inhibit the invasion and metastasis of CRC and prolong patients' disease-free survival duration. Though ECH has inhibitory activity against aggressive colorectal cancer (CRC) cells, its anti-metastasis effect in vivo and the action mechanism is undetermined. Given that ECH has an extremely low bioavailability and gut microbiota drives the CRC progression, we hypothesized that ECH could inhibit metastatic CRC by targeting the gut microbiome. AIM OF THE STUDY The purpose of this study was to investigate the impact of ECH on colorectal cancer liver metastasis in vivo and its potential mechanisms. MATERIALS AND METHODS An intrasplenic injection-induced liver metastatic model was established to examine the efficiency of ECH on tumor metastasis in vivo. Fecal microbiota from the model group and the ECH group were separately transplanted into pseudo-sterile CRLM mice in order to verify the role of gut flora in the ECH anti-metastatic effect. The 16S rRNA gene sequence was applied to analyze the structure and composition of the gut microbiota after ECH intervention, and the effect of ECH on short-chain fatty acid (SCFAs)-producing bacteria growth was proven by anaerobic culturing in vitro. GC-MS was applied to quantitatively analyze the serum SCFAs levels in mice. RNA-seq was performed to detect the gene changes involving tumor-promoting signaling pathway. RESULTS ECH inhibited CRC metastasis in a dose-dependent manner in the metastatic colorectal cancer (mCRC) mouse model. Manipulation of gut bacteria in the mCRC mouse model further proved that SCFA-generating gut bacteria played an indispensable role in mediating the antimetastatic action of ECH. Under an anaerobic condition, ECH benefited SCFA-producing microbiota without affecting the total bacterial load, presenting a dose-dependent promotion on the growth of a butyrate producer, Faecalibacterium prausnitzii (F.p). Furthermore, ECH-reshaped or F.p-colonized microbiota with a high butyrate-producing capability inhibited liver metastasis by suppressing PI3K/AKT signaling and reversing the epithelial-mesenchymal transition (EMT) process, whereas this anti-metastatic ability was abrogated by the butyrate synthase inhibitor heptanoyl-CoA. CONCLUSION This study demonstrated that ECH exhibits oral anti-metastatic efficacy by facilitating butyrate-producing gut bacteria, which downregulates PI3K/AKT signaling and EMT. It hints at a novel role for ECH in CRC therapy.
Collapse
Affiliation(s)
- Jiao Wei
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongmei Zheng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinxin Hou
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fengjing Jia
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fuwen Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng He
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ling Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
Liang H, Yin G, Shi G, Liu Z, Liu X, Li J. Echinacoside regulates PI3K/AKT/HIF-1α/VEGF cross signaling axis in proliferation and apoptosis of breast cancer. Anal Biochem 2024; 684:115360. [PMID: 37865269 DOI: 10.1016/j.ab.2023.115360] [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: 06/29/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
CONTEXT Echinacoside (ECH) is a natural anti-cancer compound and is of great value in cancer treatment. However, the mechanism underlying this effect on breast cancer (BC) was unclear. OBJECTIVE To explore the mechanism of ECH treating BC by network pharmacology and experimental validation. MATERIALS & METHODS Several databases were searched to screen potential targets of ECH and obtain information on targets related to BC. STRING was applied to construct a Protein-protein interaction (PPI) network. DAVID was applied for Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Gene Expression Profiling Interactive Analysis (GEPIA) was searched for the relationship between the expression profile and overall survival of major targets in normal breast and BC tissues. Finally, the results of network pharmacology analysis were validated by experiments. RESULTS Seventeen targets of ECH overlapped with targets in BC. Ten hub targets were determined through PPI. By GO and KEGG analysis 15 entries and 25 pathways were obtained, in which phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT), hypoxia inducible factor-1 (HIF-1) and vascular endothelial growth factor (VEGF) played greater roles. Validation of key targets in the GEPIA database showed that PIK3R1 and PIK3CD remained consistent with the results of the study. Experiments in vitro showed ECH inhibited proliferation, induced apoptosis and reduced mRNA levels and protein expression of PI3K, AKT, hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA) in MCF-7 cells. Furthermore, experiments in vivo revealed that ECH significantly reduced tumor growth, promoted apoptosis and decreased the related mRNA levels and protein expression, suggesting ECH works on BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. DISCUSSION & CONCLUSION In summary, ECH played an important role in anti-BC by regulating PI3K/AKT/HIF-1α/VEGF signaling pathway. Furthermore, ECH had multi-target and multi-pathway effects, which may be a promising natural compound for treating BC.
Collapse
Affiliation(s)
- Hongyi Liang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Guangxi Shi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Zhiyong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Xiaofei Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| | - Jingwei Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
11
|
Wang W, Jiang S, Zhao Y, Zhu G. Echinacoside: A promising active natural products and pharmacological agents. Pharmacol Res 2023; 197:106951. [PMID: 37804927 DOI: 10.1016/j.phrs.2023.106951] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Echinacoside, a natural phenylethanoid glycoside, was discovered and isolated from the garden plant Echinacea angustifolia DC., belonging to the Compositae family, approximately sixty years ago. Extensive investigations have revealed that it possesses a wide array of pharmacologically beneficial activities for human health, particularly notable for its neuroprotective and anticancer activity. Several crucial concerns surfaced, encompassing the recognition of active metabolites that exhibited inadequate bioavailability in their prototype form, the establishment of precise molecular signal pathways or targets associated with the aforementioned effects of echinacoside, and the scarcity of dependable clinical trials. Hence, the question remains unanswered as to whether scientific research can effectively utilize this natural compound. To support future studies on this natural product, it is imperative to provide a systematic overview and insights into potential future prospects. The current review provides a comprehensive analysis of the existing knowledge on echinacoside, encompassing its wide distribution, structural diversity and metabolism, diverse therapeutic applications, and improvement of echinacoside bioavailability for its potential utilization.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujun Jiang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
12
|
Alshehri A, Albuhayri A, Alanazi M, Althubaiti MA, Aljehani RF, Alsharif FI, Alatawi TM, Albalawi SS, Khodir AE, Al-Gayyar MM. Effects of Echinacoside on Ehrlich Carcinoma in Rats by Targeting Proliferation, Hypoxia and Inflammation. Cureus 2023; 15:e46800. [PMID: 37822691 PMCID: PMC10564261 DOI: 10.7759/cureus.46800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 10/13/2023] Open
Abstract
Background and objectives Ehrlich solid carcinoma (ESC) is a type of tumor originating from a spontaneous mammary adenocarcinoma in mice. It is highly aggressive and fast-growing and can create a solid undifferentiated mass when inserted under the skin. This makes it an ideal model for assessing cancer biology and tumor immunology. Echinacoside is a natural phenylethanoid glycoside with anti-inflammatory, anti-endoplasmic reticulum stress, anti-oxidative stress, and other beneficial properties. This study explored the potential anti-cancer benefits of echinacoside in rats with ESC. The study also analyzed its effects on tumor cell proliferation, differentiation, motility, and inflammation. Methods The study involved injecting rats with tumors in their left hind limb using an intramuscular injection of 2×106 cells. After 14 days, some rats were given a daily intraperitoneal dose of 30 mg/kg echinacoside for three weeks. Muscle samples were then analyzed under an electron microscope. In addition, gene expression and protein levels of various factors such as phosphoinositide 3-kinases (PI3K), mammalian target of rapamycin (mTOR), hypoxia-inducible factor (HIF)-1α, cyclin D1, cyclin-dependent kinase 2 (CDK2), tumor necrosis factor (TNF)-α, and nuclear factor (NF)κB were evaluated in another part of the muscle samples. Results After being treated with echinacoside, the ESC rats experienced a significant increase in their mean survival time from 27 days to 48 days. This treatment also resulted in a decrease in the volume and weight of the tumor. Upon examining the tumor tissue under an electron microscope, signs of damage such as pleomorphic cells, necrosis, nuclear fragmentation, membrane damage with cytoplasmic content spilling, and loss of cellular junction were observed. However, the treatment with echinacoside was effective in improving these effects. Furthermore, the expression of PI3K, mTOR, HIF-1α, cyclin D1, CDK2, TNF-α, and NFκB was significantly reduced due to the echinacoside treatment. Conclusions Our research found that echinacoside has antitumor properties that resulted in a substantial decrease in tumor size and weight, leading to an increase in the average survival time of rats and an improvement in muscle structure. Additionally, echinacoside was shown to ameliorate hypoxia by suppressing HIF-1α, reduce inflammation by decreasing NFκB and TNF-α, decrease proliferation by reducing PI3K, and block cyclin D1 and CDK2 to inhibit differentiation.
Collapse
Affiliation(s)
- Afnan Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | | | - May Alanazi
- PharmD Program, University of Tabuk, Tabuk, SAU
| | | | - Raghad F Aljehani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | - Fai I Alsharif
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | | | - Shouq S Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | - Ahmed E Khodir
- Pharmacology and Toxicology, Horus University, New Damietta, EGY
| | - Mohammed M Al-Gayyar
- Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
- Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, EGY
| |
Collapse
|
13
|
Feng D, Zhou SQ, Zhou YX, Jiang YJ, Sun QD, Song W, Cui QQ, Yan WJ, Wang J. Effect of total glycosides of Cistanche deserticola on the energy metabolism of human HepG2 cells. Front Nutr 2023; 10:1117364. [PMID: 36814512 PMCID: PMC9939456 DOI: 10.3389/fnut.2023.1117364] [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: 12/06/2022] [Accepted: 01/09/2023] [Indexed: 02/09/2023] Open
Abstract
To study the anti-tumor effect of Cistanche deserticola Y. Ma, HepG2 cells were treated with 0, 3.5, 10.5, 21, 31.5, and 42 μg/ml of total glycosides (TG) from Cistanche deserticola. The HepG2 cell survival rate and 50% inhibition concentration (IC50) were detected using the CCK-8 method, and the level of reactive oxygen species (ROS) was detected by using a DCFH-DA fluorescence probe. Finally, a Seahorse XFe24 energy analyzer (Agilent, United States) was used to detect cell mitochondrial pressure and glycolytic pressure. The results showed that TG could reduce the survival rate of HepG2 cells and that the IC50 level was 35.28 μg/ml. With increasing TG concentration, the level of ROS showed a concentration-dependent upward trend. Energy metabolism showed that each dose group of TG could significantly decline the mitochondrial respiratory and glycolytic functions of HepG2 cells. In conclusion, TG could significantly inhibit the mitochondrial respiration and glycolysis functions of HepG2 cells, increase the level of ROS, and inhibit cell proliferation. Thus, this experiment pointed out that Cistanche deserticola can be used as a source of anti-cancer foods or drugs in the future. However, further studies on its mechanisms and clinical applications are needed.
Collapse
Affiliation(s)
- Duo Feng
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Shi-qi Zhou
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Ya-xi Zhou
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Yong-jun Jiang
- Inner Mongolia Sankou Biotechnology Co., Ltd., Ordos City, Inner Mongolia, China
| | - Qiao-di Sun
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Wei Song
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Qian-qian Cui
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Wen-jie Yan
- College of Biochemical Engineering, Beijing Union University, Beijing, China
- Beijing Key Laboratory of Bioactive Substances and Functional Food, College of Biochemical Engineering, Beijing Union University, Beijing, China
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
14
|
Shi S, Qin Y, Chen D, Deng Y, Yin J, Liu S, Yu H, Huang H, Chen C, Wu Y, Zou D, Wang Z. Echinacoside (ECH) suppresses proliferation, migration, and invasion of human glioblastoma cells by inhibiting Skp2-triggered epithelial-mesenchymal transition (EMT). Eur J Pharmacol 2022; 932:175176. [PMID: 35995211 DOI: 10.1016/j.ejphar.2022.175176] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Echinacoside (ECH) is a phenylethanoid extracted from the stems of Cistanches salsa, an herb used in Chinese medicine formulations, and is effective against glioblastoma multiforme (GBM). Epithelial-mesenchymal transition (EMT) is the cornerstone of tumorigenesis and metastasis, and increases the malignant behavior of GBM cells. The S phase kinase-related protein 2 (skp2), an oncoprotein associated with EMT, is highly expressed in GBM and significantly associated with drug resistance, tumor grade and dismal prognosis. The aim of this study was to explore the inhibitory effects of ECH against GBM development and skp2-induced EMT. METHODS CCK-8, EdU incorporation, transwell, colony formation and sphere formation assays were used to determine the effects of ECH on GBM cell viability, proliferation, migration and invasion in vitro. The in vivo anti-glioma effects of ECH were examined using a U87 xenograft model. The expression levels of skp2 protein, EMT-associated markers (vimentin and snail) and stemness markers (Nestin and sox2) were analyzed by immunohistochemistry, immunofluorescence staining and western blotting experiments. RESULTS ECH suppressed the proliferation, invasiveness and migration of GBM cells in vitro, as well as the growth of U87 xenograft in vivo. In addition, ECH downregulated the skp2 protein, EMT-related markers (vimentin and snail) and stemness markers (sox2 and Nestin). The inhibitory effects of ECH were augmented in the skp2-knockdown GBM cells, and reversed in cells with ectopic expression of skp2. CONCLUSION ECH inhibits glioma development by suppressing skp2-induced EMT of GBM cells.
Collapse
Affiliation(s)
- Shengying Shi
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Yixin Qin
- Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530201, China
| | - Danmin Chen
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yanhong Deng
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Jinjin Yin
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Shaozhi Liu
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Hang Yu
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Hanhui Huang
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Chaoduan Chen
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Yinyue Wu
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Duan Zou
- Department of Pharmacy, Biomedicine Research Center, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Zhaotao Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
15
|
Shu W, Wang Z, Zhao R, Shi R, Zhang J, Zhang W, Wang H. Exploration of the Effect and Potential Mechanism of Echinacoside Against Endometrial Cancer Based on Network Pharmacology and in vitro Experimental Verification. Drug Des Devel Ther 2022; 16:1847-1863. [PMID: 35734366 PMCID: PMC9208491 DOI: 10.2147/dddt.s361955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/07/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Wan Shu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Ziwei Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Rong Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Rui Shi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Correspondence: Hongbo Wang, Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China, Email
| |
Collapse
|
16
|
Chen S, Zeng J, Huang L, Peng Y, Yan Z, Zhang A, Zhao X, Li J, Zhou Z, Wang S, Jing S, Hu M, Li Y, Wang D, Wang W, Yu H, Miao J, Li J, Deng Y, Li Y, Liu T, Xu D. RNA adenosine modifications related to prognosis and immune infiltration in osteosarcoma. J Transl Med 2022; 20:228. [PMID: 35568866 PMCID: PMC9107650 DOI: 10.1186/s12967-022-03415-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 04/27/2022] [Indexed: 11/14/2022] Open
Abstract
Background RNA adenosine modifications, which are primarily mediated by “writer” enzymes (RMWs), play a key role in epigenetic regulation in various biological processes, including tumorigenesis. However, the expression and prognostic role of these genes in osteosarcoma (OS) remain unclear. Methods Univariate and multivariate Cox analyses were used to construct the RMW signature for OS using Target datasets. RMW expression in OS tissue was detected by qPCR analysis. Xcell and GSVA were used to determine the relationship between RMWs and immune infiltration. The DGIdb and CMap databases were used for drug prediction. In vivo and in vitro experiments showed that strophanthidin elicited antitumor activity against OS. Results A 3-RMW (CSTF2, ADAR and WTAP) prognostic signature in OS was constructed using the Target dataset and verified using GEO datasets and 63 independent OS tissues via qPCR analysis. High-risk OS patients had poor overall survival, and the prognostic signature was an independent prognostic factor for OS. Functional studies showed that tumour-, metabolism-, cell cycle- and immune-related pathways were related to high risk. Next, we found that RMW-derived high-risk patients exhibited increased infiltration of M2 macrophages and cDCs. Furthermore, we predicted the potential drugs for OS using the DGIdb and CMap databases. In vivo and in vitro experiments showed that strophanthidin elicited antitumor activity against OS by repressing cell growth and inducing cell cycle arrest at the G1 phase. Conclusion The 3-RWM-based prognostic signature established in this study is a novel gene signature associated with immune infiltration, and strophanthidin was identified as a candidate therapy for OS by repressing OS cell growth and the cell cycle. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03415-6.
Collapse
Affiliation(s)
- Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Rd, Shanghai, 200241, China
| | - Jin Zeng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Liping Huang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Yi Peng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Zuyun Yan
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Aiqian Zhang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, 138, Tongzipo Road, Changsha, 410013, China
| | - Xingping Zhao
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, 138, Tongzipo Road, Changsha, 410013, China
| | - Jun Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Rd, Hefei, 230601, Anhui, China
| | - Ziting Zhou
- The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Sidan Wang
- The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Shengyu Jing
- The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Minghua Hu
- Department of Anatomy, Histology, and Embryology, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Yuezhan Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Dong Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Weiguo Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Haiyang Yu
- School of Basic Medical Science, Central South University, 172 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Jinglei Miao
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Jinsong Li
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, 138 Tongzipo Rd, Changsha, 410013, Hunan, China
| | - Yusheng Li
- Department of Orthopeadics, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, Hunan, China.
| | - Tang Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Rd, Changsha, 410011, Hunan, China.
| | - Dabao Xu
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, 138, Tongzipo Road, Changsha, 410013, China.
| |
Collapse
|
17
|
Li D, Li Q. MicroRNA-200b-3p restrains gastric cancer cell proliferation, migration, and invasion via C-X-C motif chemokine ligand 12/CXC chemokine receptor 7 axis. Bioengineered 2022; 13:6509-6520. [PMID: 35226830 PMCID: PMC8974025 DOI: 10.1080/21655979.2022.2034585] [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] [Indexed: 12/21/2022] Open
Abstract
This study was conducted to investigate the impact of microRNA (miR)-200b-3p on viability, migration, and invasion of gastric cancer (GC) cells and its mechanism. Quantitative real-time PCR (qRT-PCR) was conducted to measure miR-200b-3p expression in GC tissues and cells; besides, the relationship between miR-200b-3p expression and overall survival time (OS) was analyzed with OncomiR database; cell counting kit-8 (CCK-8), colony formation assay, flow cytometry, scratch healing assay, and Transwell assay were performed to detect the proliferation, cell cycle progression, migration, and invasion of GC cells; a lung metastasis model in nude mice was used to examine the effect of miR-200b-3p on the metastasis of GC cells in vivo; the interplay between miR-200b-3p and C-X-C motif chemokine ligand 12 (CXCL12) mRNA 3’ UTR was predicted by bioinformatics and verified with a dual-luciferase reporter gene assay; besides, the expression of CXCL12 and CXC chemokine receptor 7 (CXCR7) was probed by Western blot. It was found that miR-200b-3p expression was down-regulated in GC tissues, which was remarkably associated with the lymph node metastasis and decrease of differentiation of GC; transfection with miR-200b-3p mimics restrained the growth, migration, and invasion of GC cells in vitro, induced cell cycle arrest, and inhibited CXCL12 and CXCR7 expression levels; transfection of miR-200b-3p inhibitors worked oppositely in vitro and promoted lung metastasis in vivo. CXCL12 was confirmed as the downstream target of miR-200b-3p and was negatively modulated by miR-200b-3p. In conclusion, miR-200b-3p inhibited GC progression via regulating CXCL12/CXCR7 axis.
Collapse
Affiliation(s)
- Dinuo Li
- Department of General Gastropathy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Qiang Li
- Department of Gastrosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
18
|
Wang X, Guo S, Zhou X, Wang Y, Zhang T, Chen R. Exploring the Molecular Mechanism of lncRNA-miRNA-mRNA Networks in Non-Syndromic Cleft Lip with or without Cleft Palate. Int J Gen Med 2021; 14:9931-9943. [PMID: 34938111 PMCID: PMC8687630 DOI: 10.2147/ijgm.s339504] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/01/2021] [Indexed: 11/23/2022] Open
Abstract
Background Non-syndromic cleft lip with or without cleft palate (NSCL/P) is a common craniofacial birth defect. Growing evidence has demonstrated the competing endogenous RNA (ceRNA) hypothesis has played a role in the pathogenesis of NSCL/P. Here, we identified the important lncRNAs in NSCL/P and constructed a ceRNA regulatory network to predict their underlying functional mechanism. Methods Total RNA isolated from the peripheral blood samples were analyzed by the Human Clariom D Affymetrix platform and differentially expressed genes (DEGs) were identified. Using the limma package in R software, DEGs in the expression profile of GSE42589 were identified from Gene Expression Omnibus (GEO) database. Co-differentially expressed lncRNAs (co-DElncRNAs) were used to predict the microRNAs that may bind to them. Co-differentially expressed mRNAs (co-DEmRNAs) were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. The hub genes were screened using the cytohubba plug-in in Cytoscape. A ceRNA network was built to investigate the molecular mechanism underlying the etiology of NSCL/P. The expression levels of lncRNAs, miRNAs, and mRNAs in the network were assessed by quantitative real-time polymerase chain reaction (qRT-PCR). Results We found 116 DElncRNAs and 2955 DEmRNAs from the GSE42589 dataset, and 2626 DElncRNAs and 2771 DEmRNAs from the Human Clariom D gene chip. A network of co-DEmRNAs containing 3712 edges and 621 nodes were identified by PPI analysis. A ceRNA regulatory network comprising lncRNA USP17L6P, hsa-miR-449c-5p, and MYC was established. qRT-PCR results revealed significantly lower expression levels of lncRNA USP17L6P and c-Myc in NSCL/P tissues, while the expression level of hsa-miR-449c-5p was higher as compared to control samples (p < 0.05). Conclusion The identified lncRNAs and the established ceRNA regulatory network provide novel insight into the pathogenesis of NSCL/P, therefore hold great promise in NSCL/P management in clinical practice.
Collapse
Affiliation(s)
- Xiangpu Wang
- Department of Oral and Maxillofacial Plastic and Trauma Surgery, Center of Cleft Lip and Palate Treatment, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Siyuan Guo
- Department of Oral and Maxillofacial Plastic and Trauma Surgery, Center of Cleft Lip and Palate Treatment, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xinli Zhou
- Department of Oral and Maxillofacial Plastic and Trauma Surgery, Center of Cleft Lip and Palate Treatment, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yupei Wang
- Department of Oral and Maxillofacial Plastic and Trauma Surgery, Center of Cleft Lip and Palate Treatment, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ting Zhang
- Department of Oral and Maxillofacial Plastic and Trauma Surgery, Center of Cleft Lip and Palate Treatment, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Renji Chen
- Department of Oral and Maxillofacial Plastic and Trauma Surgery, Center of Cleft Lip and Palate Treatment, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
19
|
Concern about: Echinacoside exerts anti-tumor activity via the miR-503-3p/TGF-β1/Smad aixs in liver cancer. Cancer Cell Int 2021; 21:617. [PMID: 34809616 PMCID: PMC8609827 DOI: 10.1186/s12935-021-02311-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/01/2021] [Indexed: 11/10/2022] Open
Abstract
We concentrated on a paper in Cancer Cell International "Echinacoside exerts anti-tumor activity via the miR-503-3p/TGF-β1/Smad aixs in liver cancer". Echinacoside may be a safe and effective anti-tumor agent for the treatment of liver cancer. However, some problems in this paper made us confused.
Collapse
|