1
|
Li A, Pan W, Zhang Z, Yang F, Gou Y, Zhang Y, Ma L. Hydrazone copper(II) complexes suppressed lung adenocarcinoma by activating multiple anticancer pathway. Bioorg Chem 2025; 154:107994. [PMID: 39603071 DOI: 10.1016/j.bioorg.2024.107994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Activating multiple anti-cancer pathways has great potential for tumor treatment. Herein, we synthesized two binuclear Cu(II) hydrazone complexes ([Cu2(HL1)2Cl2] 1 and [Cu2(HL1)2Br2] 2) and two mononuclear hydrazone-Cu(II) complexes ([Cu(HL2)Cl]·CH3OH 3 and [Cu(HL2)(H2O)Br]·2H2O 4), to evaluate their anti-lung cancer activities. MTT assays revealed that the Cu(II) complexes demonstrate superior anticancer activity compared to cisplatin. Among them, complex 3 exhibited selective toxicity towards A549 cancer cells in comparison to normal cells and demonstrated hemolytic activity comparable to cisplatin. The low toxicity and effective antitumor capabilities of complex 3 have been confirmed in xenograft experiments using A549 tumor-bearing mice. Interestingly, complex 3 eradicates lung tumor cells both in vivo and in vitro by initiating multiple anticancer pathways, including cuproptosis. Our research extends the study of hydrazone copper complexes and provides strategies for the treatment of lung cancer.
Collapse
Affiliation(s)
- Aili Li
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, China
| | - Weiping Pan
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - ZhenLei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, China
| | - Yi Gou
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China.
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, China.
| | - Libing Ma
- The Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Guilin, Guangxi, China.
| |
Collapse
|
2
|
Hou X, Bai Z, Chen Y, Shi W, Yang H, Li R, Zhan X, Liu Y, Zhao X, Xiao X. Dipeptide PA3264 derived from rare and endangered Squama Manis is a novel bioactive peptide for the treatment of triple-negative breast cancer. Chin Med 2024; 19:112. [PMID: 39169391 PMCID: PMC11340106 DOI: 10.1186/s13020-024-00979-x] [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: 02/27/2024] [Accepted: 08/04/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Squama Manis is a valuable traditional Chinese medicine with a long history of medicinal use in the treatment of breast-related diseases. However, owing to the excessive exploitation and utilization of the resources, Squama Manis has been included in the list of rare and endangered wild animals. The conservation of the resources of Squama Manis and continuing its clinical application has become an urgent problem, and the search for small-molecule substitutes for Squama Manis is an effective way to achieve this goal. Previous studies have identified PA3264 as a possible active ingredient in Squama Manis. In this study, we systematically investigated the pharmacological effects and mechanisms of PA3264 in the treatment of triple-negative breast cancer (TNBC), a representative breast-related disease. METHODS Cell viability and colony formation assays were performed after treatment with the target dipeptide PA3264 in vitro. Next, 4T1 orthotopic tumors and humanized PBMC-CDX mouse models were generated to examine the antitumor effect of PA3264 in vivo. Transcriptome sequencing and molecular docking experiments were performed to predict pathways to function. Western blotting and quantitative real-time PCR were used to validate the molecular mechanisms underlying the anticancer effects of PA3264. RESULTS PA3264 significantly inhibited cell viability and migration of breast cancer cells in vitro. Furthermore, PA3264 suppressed the tumor size and reduced the tumor weight in vivo. Finally, it was verified that PA3264 prevented the progression of breast cancer by inhibiting the PI3K/AKT/NF-κB pathway, causing cell cycle arrest, and promoting apoptosis. CONCLUSIONS This study elucidated that PA3264 derived from rare and endangered Squama Manis was a novel bioactive peptide for treating triple-negative breast cancer from a scientific research perspective.
Collapse
Affiliation(s)
- Xiaorong Hou
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhaofang Bai
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yuanyuan Chen
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Huijie Yang
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Ruisheng Li
- Research Institute of Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaoyan Zhan
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Youping Liu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xu Zhao
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiaohe Xiao
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
3
|
Deng L, Ouyang B, Tang W, Wang N, Yang F, Shi H, Zhang Z, Yu H, Chen M, Wei Y, Dong J. Icariside II modulates pulmonary fibrosis via PI3K/Akt/β-catenin pathway inhibition of M2 macrophage program. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155687. [PMID: 38759312 DOI: 10.1016/j.phymed.2024.155687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/05/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a debilitating interstitial lung disorder characterized by its limited therapeutic interventions. Macrophages, particularly the alternatively activated macrophages (M2 subtype), have been acknowledged for their substantial involvement in the development of pulmonary fibrosis. Hence, targeting macrophages emerges as a plausible therapeutic avenue for IPF. Icariside II (ISE II) is a natural flavonoid glycoside molecule known for its excellent anti-tumor and anti-fibrotic activities. Nevertheless, the impact of ISE II on pulmonary fibrosis and the intricate mechanisms through which it operates have yet to be elucidated. OBJECTIVE To scrutinize the impact of ISE II on the regulation of M2 macrophage polarization and its inhibitory effect on pulmonary fibrosis, as well as to delve deeper into the underlying mechanisms of its actions. METHODS The effect of ISE II on proliferation and apoptosis in RAW264.7 cells was assessed through the use of EdU-488 labeling and the Annexin V/PI assay. Flow cytometry, western blot, and qPCR were employed to detect markers associated with the M2 polarization phenotype. The anti-fibrotic effects of ISE II in NIH-3T3 cells were investigated in a co-culture with M2 macrophages. Si-Ctnnb1 and pcDNA3.1(+)-Ctnnb1 plasmid were used to investigate the mechanism of targeted intervention. The murine model of pulmonary fibrosis was induced by intratracheal administration of bleomycin (BLM). Pulmonary function, histopathological manifestations, lung M2 macrophage infiltration, and markers associated with pulmonary fibrosis were evaluated. Furthermore, in vivo transcriptomics analysis was employed to elucidate differentially regulated genes in lung tissues. Immunofluorescence, western blot, and immunohistochemistry were conducted for corresponding validation. RESULTS Our investigation demonstrated that ISE II effectively inhibited the proliferation of RAW264.7 cells and mitigated the pro-fibrotic characteristics of M2 macrophages, exemplified by the downregulation of CD206, Arg-1, and YM-1, Fizz1, through the inhibition of the PI3K/Akt/β-catenin signaling pathway. This impact led to the amelioration of myofibroblast activation and the suppression of nuclear translocation of β-catenin of NIH-3T3 cells in a co-culture. Consequently, it resulted in decreased collagen deposition, reduced infiltration of profibrotic macrophages, and a concurrent restoration of pulmonary function in mice IPF models. Furthermore, our RNA sequencing results showed that ISE II could suppress the expression of genes related to M2 polarization, primarily by inhibiting the PI3K/Akt and β-catenin signaling pathway. In essence, our findings suggest that ISE II holds potential as an anti-fibrotic agent by orchestrating macrophage polarization. This may have significant implications in clinical practice. CONCLUSION This study has provided evidence that ISE II exerts a significant anti-fibrotic effect by inhibiting macrophage M2 polarization through the suppression of the PI3K/Akt/β-catenin signaling pathway. These findings underscore the potential of ISE II as a promising candidate for the development of anti-fibrotic pharmaceuticals in the future.
Collapse
Affiliation(s)
- Lingling Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Boshu Ouyang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Na Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Fangyong Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenhua Zhang
- Shanghai Fifth People's Hospital, Fudan University, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Mengmeng Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Li Y, Tian M, Pires Sanches JG, Zhang Q, Hou L, Zhang J. Sorcin Inhibits Mitochondrial Apoptosis by Interacting with STAT3 via NF-κB Pathway. Int J Mol Sci 2024; 25:7206. [PMID: 39000312 PMCID: PMC11241191 DOI: 10.3390/ijms25137206] [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: 05/03/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a common tumor. Our group has previously reported that sorcin (SRI) plays an important role in the progression and prognosis of HCC. This study aims to explore the mechanism of SRI inhibiting the mitochondrial apoptosis. Bioinformatics analysis, co-IP and immunofluorescence were used to analyze the relationship between SRI and STAT3. MMP and Hoechst staining were performed to detect the effect of SRI on cell apoptosis. The expression of apoptosis-related proteins and NF-κB signaling pathway were examined by Western blot and immunohistochemistry when SRI overexpression or underexpression in vivo and in vitro were found. Moreover, inhibitors were used to further explore the molecular mechanism. Overexpression of SRI inhibited cell apoptosis, which was attenuated by SRI knockdown in vitro and in vivo. Moreover, we identified that STAT3 is an SRI-interacting protein. Mechanistically, SRI interacts with STAT3 and then activates the NF-κB signaling pathway in vitro and in vivo. SRI interacting with STAT3 inhibits apoptosis by the NF-κB pathway and further contributes to the proliferation in HCC, which offers a novel clue and a new potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Yizi Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Manlin Tian
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jaceline Gislaine Pires Sanches
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Li Hou
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Jun Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
5
|
Uddin J, Fatima M, Riaz A, Kamal GM, Muhsinah AB, Ahmed AR, Iftikhar R. Pharmacological potential of micheliolide: A focus on anti-inflammatory and anticancer activities. Heliyon 2024; 10:e27299. [PMID: 38496875 PMCID: PMC10944196 DOI: 10.1016/j.heliyon.2024.e27299] [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: 12/24/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024] Open
Abstract
Micheliolide (MCL) is a chief constituent of plants such as Magnolia grandiflora L., Michelia compressa (Maxim.) Sarg. and Michelia champaca L. It is known to exhibit significant anticancer activity by various scientific investigations. This review aims to emphasize the anticancer and antiinflammatory activities of MCL. In this review, we summarized the published data in peer-reviewed manuscripts published in English. Our search was based on the following scientific search engines and databases: Scopus, Google Scholar, ScienceDirect, Springer, PubMed, and SciFinder, MCL possesses a broad spectrum of medicinal properties like other sesquiterpene lactones. The anticancer activity of this compound may be attributed to the modulation of several signaling cascades (PI3K/Akt and NF-κB pathways). It also induces apoptosis by arresting the cell cycle at the G1/G0 phase, S phase, and G2/M phase in many cancer cell lines. Very little data is available on its modulatory action on other signaling cascades like MAPK, STAT3, Wnt, TGFβ, Notch, EGFR, etc. This compound can be potentiated as a novel anticancer drug after thorough investigations in vitro, in vivo, and in silico-based studies.
Collapse
Affiliation(s)
- Jalal Uddin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Asir, 61421, Saudi Arabia
| | - Mehwish Fatima
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Ammara Riaz
- Department of Life Sciences, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, 64200, Pakistan
| | - Ghulam Mustafa Kamal
- Institute of Chemistry, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, 64200, Pakistan
| | - Abdullatif Bin Muhsinah
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Asir, 61421, Saudi Arabia
| | - Abdul Razzaq Ahmed
- Department of Prosthodontics, College of Dentistry, King Khalid University, Abha, 61421, Saudi Arabia
| | - Ramsha Iftikhar
- School of Chemistry, University of New South Wales, 2033, Sydney, Australia
| |
Collapse
|
6
|
Maqbool MF, Gul S, Ishaq M, Maryam A, Khan M, Shakir HA, Irfan M, Li Y, Ma T. Theabrownin: a dietary nutraceutical with diverse anticancer mechanisms. Nat Prod Res 2024:1-17. [PMID: 38284642 DOI: 10.1080/14786419.2024.2306917] [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: 09/14/2023] [Accepted: 01/14/2024] [Indexed: 01/30/2024]
Abstract
Cancer, a highly deadly disease, necessitates safe, cost-effective, and readily accessible treatments to mitigate its impact. Theabrownin (THBR), a polyphenolic pigment found in Pu-erh tea, has garnered attention for its potential benefits in memory, liver health, and inflammation control. By observing different biological activities of THBR, recently researchers have unveiled THBR's promising anticancer properties across various human cancer types. By examining existing studies, it is evident that THBR demonstrates substantial potential in inhibiting cell proliferation and reducing tumour size with minimal harm to normal cells. These effects are achieved through the modulation of key molecular markers such as Bcl-2, Bax, various Caspases, Poly (ADP-ribose) polymerase cleavage (Cl-PARP), and zinc finger E box binding homeobox 1 (ZEB 1). This review aims to provide in-depth insights into THBR's role in cancer research. This review also elucidates the underlying anticancer mechanisms of THBR, offering promise as a novel anticancer drug to alleviate the global cancer burden.
Collapse
Affiliation(s)
- Muhammad Faisal Maqbool
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Sameena Gul
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Ishaq
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Amara Maryam
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Khan
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Hafiz Abdullah Shakir
- Cancer Research Laboratory, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Yongming Li
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tonghui Ma
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
7
|
Zhao Z, Du JF, Wang QL, Qiu FN, Li P, Jiang Y, Li HJ. Natural Product Baohuoside I Impairs the Stability and Membrane Location of MRP2 Reciprocally Regulated by SUMOylation and Ubiquitination in Hepatocytes. Chem Res Toxicol 2024; 37:57-71. [PMID: 38177062 DOI: 10.1021/acs.chemrestox.3c00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
Epimedii Folium (EF) is a botanical dietary supplement to benefit immunity. Baohuoside I (BI), a prenylated flavonoid derived from EF, has exhibited the cholestatic risk before. Here, the mechanism of BI on the stability and membrane localization of liver MRP2, a bile acid exporter in the canalicular membrane of hepatocytes, was investigated. The fluorescent substrate of MRP2, CMFDA was accumulated in sandwich-cultured primary mouse hepatocytes (SCH) under BI stimulation, followed by reduced membrane MRP2 expression. BI triggered MRP2 endocytosis associated with oxidative stress via inhibition of the NRF2 signaling pathway. Meanwhile, BI promoted the degradation of MRP2 by reducing its SUMOylation and enhancing its ubiquitination level. Co-IP and fluorescence colocalization experiments all proved that MRP2 was a substrate protein for SUMOylation for SUMO proteins. CHX assays showed that SUMO1 prolonged the half-life of MRP2 and further increased its membrane expression, which could be reversed by UBC9 knockdown. Correspondingly, MRP2 accumulated in the cytoplasm by GP78 knockdown or under MG132 treatment. Additionally, the SUMOylation sites of MRP2 were predicted by the algorithm, and a conversion of lysines to arginines at positions 940 and 953 of human MRP2 caused its decreased stability and membrane location. K940 was further identified as the essential ubiquitination site for MRP2 by an in vitro ubiquitination assay. Moreover, the decreased ubiquitination of MRP2 enhanced the SUMOylation MRP2 and vice versa, and the crosstalk of these two modifiers could be disrupted by BI. Collectively, our findings indicated the process of MRP2 turnover from the membrane to cytoplasm at the post-translational level and further elucidated the novel toxicological mechanism of BI.
Collapse
Affiliation(s)
- Zhen Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jin-Fa Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Qiao-Lei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Fang-Ning Qiu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yan Jiang
- College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| |
Collapse
|
8
|
Wang T, Zhang M, Khan M, Li J, Wu X, Ma T, Li Y. Cryptotanshinone suppresses ovarian cancer via simultaneous inhibition of glycolysis and oxidative phosphorylation. Biomed Pharmacother 2024; 170:115956. [PMID: 38039759 DOI: 10.1016/j.biopha.2023.115956] [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: 07/14/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023] Open
Abstract
Ovarian cancer is one of the most lethal cancers in female reproductive system due to heterogeneity and lack of effective treatment. Targeting aerobic glycolysis, a predominant energy metabolism of cancer cells has been recognized a novel strategy to overcome cancer cell growth. However, the capability of cancer cells to undergo metabolic reprogramming guarantees their survival even when glycolysis is inhibited. Here in this study, we have shown that Cryptotanshinone (CT), a lipid-soluble bioactive anticancer molecule of Salvia miltiorrhiza, inhibits both glycolysis and oxidative phosphorylation (OXPHOS) in ovarian cancer cells leading to growth suppression and apoptosis induction. Our mechanistic study revealed that CT decreased glucose uptake and lactate production, and inhibited the kinase activity of LDHA and HK2. The molecular docking study showed that CT could directly bind with GLUT1, LDHA, HK2, PKM2 and complex-1. The immunoblotting data showed that CT decreased the expression of aberrantly activated glycolytic proteins includingGLUT1, LDHA, HK2, and PKM2. Besides, we found that CT inhibited mitochondrial ComplexⅠ activity, decreased the ratio of NAD+/NADH, and suppressed the generation of ATP and induced activation of AMPK, which controls energy-reducing processes. These in vitro findings were further validated using xenograft model. The findings of in vivo studies were in line with in vitro studies. Taken together, CT effectively suppressed glycolysis and OXPHOS, inhibited growth and induced apoptosis in ovarian cancer cells both in vitro and in vivo study models.
Collapse
Affiliation(s)
- Tong Wang
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengmeng Zhang
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan.
| | - Jingjing Li
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Wu
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tonghui Ma
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yongming Li
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
9
|
Zhang X, Tang B, Wen S, Wang Y, Pan C, Qu L, Yin Y, Wei Y. Advancements in the Biotransformation and Biosynthesis of the Primary Active Flavonoids Derived from Epimedium. Molecules 2023; 28:7173. [PMID: 37894651 PMCID: PMC10609448 DOI: 10.3390/molecules28207173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Epimedium is a classical Chinese herbal medicine, which has been used extensively to treat various diseases, such as sexual dysfunction, osteoporosis, cancer, rheumatoid arthritis, and brain diseases. Flavonoids, such as icariin, baohuoside I, icaritin, and epimedin C, are the main active ingredients with diverse pharmacological activities. Currently, most Epimedium flavonoids are extracted from Epimedium plants, but this method cannot meet the increasing market demand. Biotransformation strategies promised huge potential for increasing the contents of high-value Epimedium flavonoids, which would promote the full use of the Epimedium herb. Complete biosynthesis of major Epimedium flavonoids by microbial cell factories would enable industrial-scale production of Epimedium flavonoids. This review summarizes the structures, pharmacological activities, and biosynthesis pathways in the Epimedium plant, as well as the extraction methods of major Epimedium flavonoids, and advancements in the biotransformation and complete microbial synthesis of Epimedium flavonoids, which would provide valuable insights for future studies on Epimedium herb usage and the production of Epimedium flavonoids.
Collapse
Affiliation(s)
- Xiaoling Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Laboratory of Synthetic Biology, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Food Safety Quick Testing and Smart Supervision Technology for State Market Regulation, Zhengzhou 450003, China
| | - Bingling Tang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Laboratory of Synthetic Biology, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Sijie Wen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Laboratory of Synthetic Biology, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yitong Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Laboratory of Synthetic Biology, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Chengxue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lingbo Qu
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Yulong Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410081, China
| | - Yongjun Wei
- Laboratory of Synthetic Biology, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
10
|
Feng X, Yin Z, Ou S, Chu Z, Feng J, Luo Y, Hu Y, Liu Y, Jiang W, Wang X, Wang H. The anti-tumor effects of Celastrus orbiculatus Thunb. and its monomer composition: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 310:116363. [PMID: 36948266 DOI: 10.1016/j.jep.2023.116363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/04/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Celastrus orbiculatus Thunb. has been included in "The Plant List" (http://www. theplantlist.org) and is the most widely researched species in its genus. It is called Nanshe Teng in China. Celastrus orbiculatus Thunb. is a plant of Euonymus and it's medicinal part is the vine and stem. It is also called Alias Dragon grass, Yellow Yine, etc. It has good anti-tumor, anti-inflammatory and other effects. More and more studies have shown that Celastrus orbiculatus Thunb. has a significant therapeutic effect on a variety of malignant tumors. The research on Celastrus orbiculatus Thunb. has a good application prospect for the development of anti-tumor drugs. However, no systematic reports on Celastrus orbiculatus Thunb. have been published before. AIM OF THE REVIEW This paper summarizes the metabolic products for anti-tumor and the mechanism for anti-tumor of Celastrus orbiculatus Thunb. to provide reference for further development and research. MATERIALS AND METHODS The relevant information on Celastrus orbiculatus Thunb. was collected from the scientific databases including PubMed, CNKI, ScienceDirect, Wiley, Springer, Web of Science, Google Scholar, Baidu Scholar, Pharmacopoeia of the People's Republic of China and Flora Republicae Popularis Sinicae, etc. RESULTS: At present, more than 200 compounds have been identified from Celastrus orbiculatus Thunb., including terpenoids, flavonoids, phenylpropanoids, polyketides and benzene derivatives, etc. Pharmacological studies have shown that Celastrus orbiculatus Thunb. has a variety effects of inhibiting tumor cell proliferation, inducing tumor cell apoptosis, inhibiting tumor cells invasion, metastasis and angiogenesis, reversing multi-drug resistance, and also collaborativing Micro RNA to inhibit tumor growth, etc. It has a significant effect on gastric cancer, liver cancer, lung cancer, etc. The extracts of Celastrus orbiculatus Thunb. have been widely used in experiments, and the toxic and side effects are small. CONCLUSIONS Celastrus orbiculatus Thunb. is rich in chemical constituents, diverse in pharmacological activities and abundant in resources, which is widely used in clinics from traditional to modern. However, there is no systematic report on the chemical compounds and anti-tumor effects of Celastrus orbiculatus Thunb. We organize and summarize it to provide reference for further development and research.
Collapse
Affiliation(s)
- Xinyi Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Zixin Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Shiya Ou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Zewen Chu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Jun Feng
- Department of Oncology, Gaoyou Hospital of Traditional Chinese Medicine, Yangzhou, 225600, PR China.
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Yaqi Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| | - Wei Jiang
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China; College of Environmental Science & Engineering, Yangzhou University, Yangzhou, Jiangsu, 225127, China.
| | - Xiaoqing Wang
- Department of Oncology, Gaoyou Hospital of Traditional Chinese Medicine, Yangzhou, 225600, PR China.
| | - Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, 225001, China.
| |
Collapse
|
11
|
Wang T, Yin S, Gu J, Li J, Zhang M, Shan J, Wu X, Li Y. Study on the Intervention Mechanism of Cryptotanshinone on Human A2780 Ovarian Cancer Cell Line Using GC-MS-Based Cellular Metabolomics. Pharmaceuticals (Basel) 2023; 16:861. [PMID: 37375808 DOI: 10.3390/ph16060861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cryptotanshinone (CT), an active component of the traditional Chinese medicine Salvia miltiorrhiza Bunge, exhibits a wide range of biological and pharmacological activities. Although the anticancer activity of CT is well known, the knowledge of its effect on the regulation of cancer cell metabolism is relatively new. The present study investigated the anticancer mechanism of CT in ovarian cancer with a focus on cancer metabolism. CCK8 assays, apoptosis assays, and cell cycle assays were conducted to reveal the growth-suppressive effect of CT on ovarian cancer A2780 cells. To explore the potential underlying mechanisms of CT, the changes in endogenous metabolites in A2780 cells before and after CT intervention were investigated using the gas chromatography-mass spectrometry (GC-MS) approach. A total of 28 important potential biomarkers underwent significant changes, mainly involving aminoacyl-tRNA biosynthesis, energy metabolism, and other pathways. Changes in the ATP and amino acid contents were verified with in vitro and in vivo experiments. Our results indicate that CT may exert an anti-ovarian cancer effect by inhibiting ATP production, promoting the protein catabolic process, and inhibiting protein synthesis, which may lead to cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Tong Wang
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shusheng Yin
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juan Gu
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jingjing Li
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengmeng Zhang
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Wu
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
12
|
Gani I, Jameel S, Bhat SA, Amin H, Bhat KA. Prenylated Flavonoids of Genus Epimedium: Phytochemistry, Estimation and Synthesis. ChemistrySelect 2023. [DOI: 10.1002/slct.202204263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Affiliation(s)
- Ifshana Gani
- CSIR-Indian Institute of Integrative Medicine Srinagar, Jammu and Kashmir 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Salman Jameel
- CSIR-Indian Institute of Integrative Medicine Srinagar, Jammu and Kashmir 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Showkat Ahmad Bhat
- CSIR-Indian Institute of Integrative Medicine Srinagar, Jammu and Kashmir 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Henna Amin
- CSIR-Indian Institute of Integrative Medicine Srinagar, Jammu and Kashmir 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Khursheed Ahmad Bhat
- CSIR-Indian Institute of Integrative Medicine Srinagar, Jammu and Kashmir 190005 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| |
Collapse
|
13
|
Kong Q, Ma M, Zhang L, Liu S, He S, Wu J, Liu B, Dong J. Icariside II potentiates the anti-PD-1 antitumor effect by reducing chemotactic infiltration of myeloid-derived suppressor cells into the tumor microenvironment via ROS-mediated inactivation of the SRC/ERK/STAT3 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154638. [PMID: 36621167 DOI: 10.1016/j.phymed.2022.154638] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/08/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Immune checkpoint blockade agents, such as anti-PD-1 antibodies, show promising antitumor efficacy but only a limited response in patients with non-small cell lung cancer (NSCLC). Icariside II (IS), a metabolite of Herba Epimedii, is a COX-2 and EGFR inhibitor that can enhance the anti-PD-1 effect. This study aimed to evaluate the antitumor effect of IS in combination with anti-PD-1 and explore the underlying mechanism. METHODS Tumor growth was assessed in Lewis Lung Cancer (LLC) tumor-bearing mice in seven groups (control, IS 20 mg/kg, IS 40 mg/kg, anti-PD-1, IS 20 mg/kg+anti-PD-1, IS 40 mg/kg+anti-PD-1, ERK inhibitor+anti-PD-1). Tumor-infiltrating immune cells were measured by flow cytometry. The mechanisms were explored by tumor RNA-seq and validated in LLC cells through molecular biological experiments using qRT‒PCR, ELISA, and western blotting. RESULTS Animal experiments showed that IS in combination with anti-PD-1 further inhibited tumor growth and remarkably reduced the infiltration of myeloid-derived suppressor cells (MDSCs) into the tumor compared with anti-PD-1 monotherapy. RNA-seq and in vitro experiments showed that IS suppressed the chemotactic migration of MDSCs by downregulating the expression of CXC chemokine ligands 2 (CXCL2) and CXCL3. Moreover, IS promoted reactive oxygen species (ROS) generation and inhibited the activation of SRC/ERK/STAT3 in LLC cells, which are upstream signaling pathways of these chemokines. CONCLUSION IS potentiates the anti-PD-1 anti-tumor effect by reducing chemotactic infiltration of the myeloid-derived suppressor cell into the tumor microenvironment, via ROS-mediated inactivation of SRC/ERK/STAT3 signaling pathways.
Collapse
Affiliation(s)
- Qing Kong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Mengyu Ma
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Li Zhang
- Department of Neurology, Huadong Hospital, Fudan University, Shanghai, China
| | - Suqing Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shan He
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Baojun Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Gul S, Maqbool MF, Maryam A, Khan M, Shakir HA, Irfan M, Ara C, Li Y, Ma T. Vitamin K: A novel cancer chemosensitizer. Biotechnol Appl Biochem 2022; 69:2641-2657. [PMID: 34993998 DOI: 10.1002/bab.2312] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022]
Abstract
Cancer incidences are growing rapidly and causing millions of deaths globally. Cancer treatment is one of the most exigent challenges. Drug resistance is a natural phenomenon and is considered one of the major obstacles in the successful treatment of cancer by chemotherapy. Combination therapy by the amalgamation of various anticancer drugs has suggested modulating tumor response by targeting various signaling pathways in a synergistic or additive manner. Vitamin K is an essential nutrient and has recently been investigated as a potential anticancer agent. The combination of vitamin K analogs, such as vitamins K1, K2, K3, and K5, with other chemotherapeutic drugs have demonstrated a safe, cost-effective, and most efficient way to overcome drug resistance and improved the outcomes of prevailing chemotherapy. Published reports have shown that vitamin K in combination therapy improved the efficacy of clinical drugs by promoting apoptosis and cell cycle arrest and overcoming drug resistance by inhibiting P-glycoprotein. In this review, we discuss the mechanism, cellular targets, and possible ways to develop vitamin K subtypes into effective cancer chemosensitizers. Finally, this review will provide a scientific basis for exploiting vitamin K as a potential agent to improve the efficacy of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Sameena Gul
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Muhammad Faisal Maqbool
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Amara Maryam
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Hafiz Abdullah Shakir
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Chaman Ara
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus Lahore, Lahore, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
15
|
Song W, Yuan Y, Tan X, Gu Y, Zeng J, Song W, Xin Z, Fang D, Guan R. Icariside II induces rapid phosphorylation of endothelial nitric oxide synthase via multiple signaling pathways. PeerJ 2022; 10:e14192. [PMID: 36312762 PMCID: PMC9615964 DOI: 10.7717/peerj.14192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/15/2022] [Indexed: 01/24/2023] Open
Abstract
Icariside II, as a favonoid compound derived from epimedium, has been proved to involed in a variety of biological and pharmacological effects such as anti-inflammatory, anti-osteoporosis, anti-oxidation, anti-aging, and anti-cancer but its mechanism is unclear, especially in terms of its effect on post-transcriptional modification of endothelial nitric oxide synthase (eNOS). Phosphorylation of eNOS plays an important role in the synthesis of nitric oxide in endothelial cells, which is closely related to erectile dysfunction, atherosclerosis, Alzheimer's disease, and other diseases. Our study aims to investigate the effect and mechanism of Icariside II on the rapid phosphorylation of eNOS. In this study, human umbilical vein endothelial cells (HUVECs) were stimulated with Icariside II in the presence or absence of multiple inhibitors (1 µM), including LY294002 (PI3K-inhibitor), MK-2206 (AKT-inhibitor), Bisindolylmaleimide X (AMPK-inhibitor), H-89 (CaMKII-inhibitor), KN-62 (PKA-inhibitor), Dorsomorphin (PKC-inhibitor). The proliferation of HUVECs was assessed using cell counting kit-8 (CCK-8). The release of nitric oxide (NO) within HUVECs was detected via fluorescence probe (DAF-FM). Western blot was used to examine the effect of Icariside II on the expression of eNOS, phosphorylation of eNOS, and common signaling pathways proteins. In this study, Icariside II was found to promote the cell proliferation and rapid NO release in HUVECs. The phosphorylation of eNOS-Ser1177 was significantly increased after Icariside II stimulation and reached a peak at 10 min (p < 0.05). Meanwhile, the phosphorylation of eNOS-Thr495 was significantly decreased after 45 min of stimulation (p < 0.05). Following the intervention with multiple inhibitors, it was found that MK-2206 (AKT inhibitor), LY294002 (PI3K inhibitor), KN-62 (AMPK inhibitor), and Bisindolylmaleimide X (PKC inhibitor) could significantly inhibit the phosphorylation of eNOS-Ser1177 caused by Icariside II (p < 0.05), while MK-2206, LY294002, and Bisindolylmaleimide X reversed the alleviated phosphorylation of eNOS-Thr495. We concluded that Icariside can regulate rapid phosphorylation of eNOS- Ser1177 and eNOS-Thr495 via multiple signaling pathways, resulting in the up-regulation of eNOS and the increased release of NO.
Collapse
Affiliation(s)
- Wenpeng Song
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China,Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yiming Yuan
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Xiaohui Tan
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Yangyang Gu
- Department of Urology, Peking University First Hospital, Beijing, China,Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jianyu Zeng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Weidong Song
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Zhongcheng Xin
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Dong Fang
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Ruili Guan
- Department of Urology, Peking University First Hospital, Beijing, China,Institute of Urology, Peking University, Beijing, China,Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| |
Collapse
|
16
|
Rahman MM, Sarker MT, Alam Tumpa MA, Yamin M, Islam T, Park MN, Islam MR, Rauf A, Sharma R, Cavalu S, Kim B. Exploring the recent trends in perturbing the cellular signaling pathways in cancer by natural products. Front Pharmacol 2022; 13:950109. [PMID: 36160435 PMCID: PMC9498834 DOI: 10.3389/fphar.2022.950109] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/15/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is commonly thought to be the product of irregular cell division. According to the World Health Organization (WHO), cancer is the major cause of death globally. Nature offers an abundant supply of bioactive compounds with high therapeutic efficacy. Anticancer effects have been studied in a variety of phytochemicals found in nature. When Food and Drug Administration (FDA)-approved anticancer drugs are combined with natural compounds, the effectiveness improves. Several agents have already progressed to clinical trials based on these promising results of natural compounds against various cancer forms. Natural compounds prevent cancer cell proliferation, development, and metastasis by inducing cell cycle arrest, activating intrinsic and extrinsic apoptosis pathways, generating reactive oxygen species (ROS), and down-regulating activated signaling pathways. These natural chemicals are known to affect numerous important cellular signaling pathways, such as NF-B, MAPK, Wnt, Notch, Akt, p53, AR, ER, and many others, to cause cell death signals and induce apoptosis in pre-cancerous or cancer cells without harming normal cells. As a result, non-toxic “natural drugs” taken from nature’s bounty could be effective for the prevention of tumor progression and/or therapy of human malignancies, either alone or in combination with conventional treatments. Natural compounds have also been shown in preclinical studies to improve the sensitivity of resistant cancers to currently available chemotherapy agents. To summarize, preclinical and clinical findings against cancer indicate that natural-sourced compounds have promising anticancer efficacy. The vital purpose of these studies is to target cellular signaling pathways in cancer by natural compounds.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Taslim Sarker
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mst. Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Yamin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Tamanna Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Pakistan
- *Correspondence: Abdur Rauf, ; Bonglee Kim,
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Abdur Rauf, ; Bonglee Kim,
| |
Collapse
|
17
|
An Overview on Rumex dentatus L.: Its Functions as a Source of Nutrient and Health-Promoting Plant. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8649119. [PMID: 35911153 PMCID: PMC9337939 DOI: 10.1155/2022/8649119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022]
Abstract
Rumex dentatus L. (Polygonaceae), also known as toothed dock or Aegean dock, is a medicinal plant with a high culinary value in addition to being used as an ethnomedicinal plant. This review focuses on the botanical, nutritional, phytochemical, and pharmacological activities of R. dentatus, as well as the future prospects for systematic investigations into these areas. R. dentatus has been subjected to scientific evaluation, which has confirmed its traditional uses and demonstrated a wide range of biological and pharmacological potentials, including antioxidant, anticancer, antifungal, antibacterial, anti-inflammatory, and other biological properties. Phytochemical analyses showed the presence of anthraquinones, chromones, flavonoids, and essential oils. As a result of this current review, the medicinal significance of R. dentatus has been confirmed, and future research on its unexplored aspects, such as the identification of pharmacologically active chemical constituents and related mechanisms and safety, may be stimulated, with the goal of developing it into a drug.
Collapse
|
18
|
Chen G, Huang J, Lei H, Wu F, Chen C, Song Y, Cao Z, Zhang C, Zhang C, Ma Y, Huang M, Zhou J, Lu Y, Zhao Y, Zhang L. Icariside I - A novel inhibitor of the kynurenine-AhR pathway with potential for cancer therapy by blocking tumor immune escape. Biomed Pharmacother 2022; 153:113387. [PMID: 35834991 DOI: 10.1016/j.biopha.2022.113387] [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: 06/03/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Although therapeutic antibodies against immune checkpoints such as PD-1/PD-L1 have achieved unprecedented success in clinical tumor patients, there are still many patients who are ineffective or have limited responses to immune checkpoint blockade (ICB). Discovery of novel strategies for cancer immunotherapy including natural small molecules is needed. METHODS Owing to its extremely low content in Epimedium genus, we firstly constructed a microbial cell factory to enzymatically biosynthesize icariside I, a natural flavonoid monosaccharide from Herbal Epimedium. Using a combination of targeted MS-based metabolomics, flow cytometric analysis, and biological assays, the therapeutic potentials of icariside I were subsequently investigated in vivo and in vitro. RESULTS We find that icariside I markedly downregulates a series of intermediate metabolites such as kynurenine, kynurenic acid and xanthurenic acid and corresponding key enzymes involved in kynurenine-AhR pathway in both tumor cells and tumor-bearing mice. In vivo, oral administration of icariside I downregulates SLC7A8 and PAT4 transporters and AhR, thus inhibiting nuclear PD-1 in CTLs. Moreover, icariside I significantly upregulates CD8 + T cells in both peripheral blood and tumor tissues of tumor-bearing mice. Consequently, interferon-γ (IFN-γ) secreted by CD8 + T cells suppresses tumor growth through activation of JAK1-STAT1 signaling, thus inducing tumor cell apoptosis. CONCLUSIONS These results suggest that icariside I could be an effective small molecule drug for tumor immunotherapy by blocking kynurenine-AhR pathway and tumor immune escape.
Collapse
Affiliation(s)
- Gui Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiajun Huang
- Golden Health (Guangdong) Biotechnology Co., Ltd, Foshan 528225, China; School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Hehua Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China
| | - Fang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chuan Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuchen Song
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng Cao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ce Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cui Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxi Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mingtao Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Jinlin Zhou
- Golden Health (Guangdong) Biotechnology Co., Ltd, Foshan 528225, China
| | - Yujing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yanxia Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Limin Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, CAS, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
19
|
Yuan D, Guo T, Qian H, Ge H, Zhao Y, Huang A, Wang X, Cao X, Zhu D, He C, Yu H. Icariside II suppresses the tumorigenesis and development of ovarian cancer by regulating miR-144-3p/IGF2R axis. Drug Dev Res 2022; 83:1383-1393. [PMID: 35808943 DOI: 10.1002/ddr.21967] [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: 03/24/2022] [Revised: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 11/10/2022]
Abstract
Ovarian cancer is one of the three major gynecological malignancies. It has been reported that Icariside II was able to block the occurrence and development of ovarian cancer. However, the detailed mechanism by which Icariside II regulates the development of ovarian cancer is widely unknown. EdU staining and transwell assays were applied to detect the proliferation, migration, and invasion of ovarian cancer cells. Next, the relationship between miR-144-3p and IGF2R was verified by the dual-luciferase reporter assay. Moreover, in vivo animal model was constructed to verify the effect of Icariside II on the development of ovarian cancer. Icariside II notably inhibited the proliferation, migration, and invasion and induced the apoptosis of ovarian cancer cells. Additionally, Icariside II markedly increased the level of miR-144-3p in ovarian cancer cells. Moreover, IGF2R was targeted by miR-144-3p directly. Icariside II significantly decreased the expression of IGF2R and the phosphorylation level of AKT and mTOR in ovarian cancer cells, which were partially reversed by miR-144-3p inhibitor. Meanwhile, Icariside II remarkably promoted the autophagy of ovarian cancer cells, as confirmed by the increased expression of Beclin-1 and ATG-5 and decreased expression of p62; however, co-treatment with miR-144-3p inhibitor notably decreased autophagy. Furthermore, the result of animal study suggested Icariside II notably inhibited ovarian tumor growth as well. Collectively, Icariside II could suppress the tumorigenesis and development of ovarian cancer by promoting autophagy via miR-144-3p/IGF2R axis. These results may be beneficial for future studies on the use of Icariside II to treat ovarian cancer.
Collapse
Affiliation(s)
- Donglan Yuan
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Ting Guo
- Center for Molecular Medicine, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Hua Qian
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Hongshan Ge
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Yinling Zhao
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Aihua Huang
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Xiaosu Wang
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Xiuhong Cao
- Department of Operation, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - DanDan Zhu
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - CuiQin He
- Department of Obstetrics and Gynecology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| | - Hong Yu
- Department of Pathology, Taizhou People's Hospital, Affiliated Hospital of NanJing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|
20
|
Gu YY, Tan XH, Song WP, Song WD, Yuan YM, Xin ZC, Wang JD, Fang D, Guan RL. Icariside Ⅱ Attenuates Palmitic Acid-Induced Endothelial Dysfunction Through SRPK1-Akt-eNOS Signaling Pathway. Front Pharmacol 2022; 13:920601. [PMID: 35846993 PMCID: PMC9280058 DOI: 10.3389/fphar.2022.920601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Endothelial dysfunction is commonly accompanied by a reduced capacity for nitric oxide (NO) production and decreased NO sensitivity, playing a central role in numerous vascular diseases. Saturated free fatty acids are known to reduce NO production and then induce endothelial dysfunction. Alternative splicing participates in the regulation of cellular and tissular homeostasis and is highly regulated by serine-arginine protein kinase (SRPK1). The role of SRPK1 in the biology of endothelial cells remains elusive. Icariside Ⅱ (ICA Ⅱ) has been reported to have protective effects on endothelial function. However, the specific molecular mechanisms are still unknown. The purpose of this study is to explore the role of SRPK1 in the biology of endothelial cells and the underlying mechanism of ICA Ⅱ on palmitic acid (PA) induced endothelial dysfunction. Methods: Endothelial dysfunction was induced using PA in human umbilical vein endothelial cells (HUVECs). The expression and phosphorylation of related proteins in the SRPK1-Akt-eNOS signaling pathway were detected by Western Blot. Cell Counting Kit-8 assay and Ki-67 immunofluorescence were used to estimate cell viability. Endothelial cell function was assessed by detecting NO production using DAF-FM DA. Interaction between ICA Ⅱ and SRPK1 was demonstrated by a biotinylated protein interaction pull-down assay. Results: The expressions of eNOS, Akt, and SRPK1 were down-regulated in the endothelial dysfunction stimulated by PA. SRPK1 inhibitor SPHINX31 restrained endothelial cell viability in a dose-dependent manner. Moreover, inhibition of SRPK1 using SPHINX31 and knockdown of SRPK1 by shRNA also showed a down-regulation of the proteins associated with the SRPK1-Akt-eNOS signaling pathway. Biotinylated protein interaction pull-down assay revealed that ICA Ⅱ could be directly bound with SRPK1. On the other hand, ICA Ⅱ could attenuate the PA-induced endothelial dysfunction and restore cell viability through the SRPK1-Akt-eNOS pathway. Conclusions: ICA Ⅱ, bound with SRPK1, could attenuate the endothelial dysfunction induced by the PA in HUVECs via the SRPK1-Akt-eNOS signaling pathway.
Collapse
Affiliation(s)
- Yang-Yang Gu
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
| | - Xiao-Hui Tan
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Wen-Peng Song
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wei-Dong Song
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Yi-Ming Yuan
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
| | - Zhong-Cheng Xin
- Male Reproductive and Sexual Medicine, Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Institute of Urology, Tianjin Medical University, Tianjin, China
| | - Jia-Dong Wang
- Department of Radiation Medicine, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- *Correspondence: Rui-Li Guan, ; Dong Fang, ; Jia-Dong Wang,
| | - Dong Fang
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
- *Correspondence: Rui-Li Guan, ; Dong Fang, ; Jia-Dong Wang,
| | - Rui-Li Guan
- Department of Urology, Peking University First Hospital, Beijing, China
- Institute of Urology, Peking University, Beijing, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, China
- *Correspondence: Rui-Li Guan, ; Dong Fang, ; Jia-Dong Wang,
| |
Collapse
|
21
|
Ji D, Hou L, Xie C, Feng H, Bao D, Teng Y, Liu J, Cui T, Wang X, Xu Y, Tan G. Deoxyelephantopin Suppresses Pancreatic Cancer Progression In Vitro and In Vivo by Targeting linc00511/miR-370-5p/p21 Promoter Axis. JOURNAL OF ONCOLOGY 2022; 2022:3855462. [PMID: 35794978 PMCID: PMC9252706 DOI: 10.1155/2022/3855462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/30/2022] [Indexed: 12/02/2022]
Abstract
Objectives Deoxyelephantopin (DET) is a kind of natural active ingredient extracted from the Chinese herbal medicine Elephantopus scaber L. Many studies have revealed the potential antitumor effect on multiple malignancies. However, the detailed mechanism of its antitumor effect in pancreatic cancer remains unclear. Recently, studies have confirmed that noncoding RNA (ncRNA) plays an important regulatory role in malignancies. This research was performed to explore the relationship between ncRNA and DET-induced tumor inhibition in pancreatic cancer. Methods Microarray profiling was applied to identify the candidate ncRNAs associated with DET-induced tumor inhibition. Quantitative real-time PCR was used to evaluate the expression of linc00511 in pancreatic cancer cells and tissues. The influence of DET on the cell proliferation, migration, and invasion was assessed by CCK-8, colony formation, wound healing, and Transwell assays. The relationship between lncRNAs, miRNAs, and p21 promoter region was analyzed by bioinformatics and verified by luciferase reporter gene and western blotting. The effect of linc00511 on nuclear translocation of miR-370-5p was explored by cytoplasmic and nuclear RNA purification. Moreover, the effect of DET on tumor growth and metastasis, and the prophylactic effect were investigated by establishing subcutaneous and lung metastatic tumor models. Results Microarray assay indicated linc00511 was a potential target gene. The antitumor effect of DET in pancreatic cancer depended on downregulating linc00511 expression, and linc00511 might be an oncogene in pancreatic cancer. Silencing linc00511 enhanced the antitumor function of DET; conversely, linc00511 overexpression antagonized the DET cytotoxic effect. Additionally, miR-370-5p could bind to p21 promoter to exert the RNA activation and then promote p21 expression. P21 was a downstream gene of linc00511 and associated with pancreatic cancer progression. Linc00511 regulated p21 expression by blocking miR-370-5p nuclear translocation. Conclusions To sum up, the present finding confirmed that DET suppressed the malignant biological behavior of pancreatic cancer via linc00511/miR-370-5p/p21 promoter axis.
Collapse
Affiliation(s)
- Daolin Ji
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
| | - Li Hou
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chunyang Xie
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Haonan Feng
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Dongdong Bao
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yue Teng
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Junhao Liu
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tiangang Cui
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiuhong Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Gang Tan
- Department of Hepatopancreatobiliary Surgery, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
22
|
Laka K, Mbita Z. P53-Related Anticancer Activities of Drimia calcarata Bulb Extracts Against Lung Cancer. Front Mol Biosci 2022; 9:876213. [PMID: 35769912 PMCID: PMC9235921 DOI: 10.3389/fmolb.2022.876213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/02/2022] [Indexed: 01/18/2023] Open
Abstract
Current lung cancer treatment strategies are ineffective, and lung cancer cases continue to soar; thus, novel anticancer drugs and targets are needed, and medicinal plants are promising to offer better alternatives. This study was aimed at analysing two p53 splice variants during the potential anticancer activities of Drimia calcarata (Dc) methanol and water extracts against different human lung cancer cell lines of varying p53 mutation status, and these included mutant H1573 and mutant H1437 and p53-wild type (A549) cells. The anticancer activities of the Dc extracts were assessed by establishing the cytotoxic effect and the apoptosis-inducing capacity of these extracts, using the MTT assay and Annexin V analysis, respectively, with the latter confirmed using fluorescence microscopy. The molecular mechanisms induced by these extracts were further evaluated using cell cycle analysis and RT-PCR. Both extracts demonstrated safety against noncancerous lung MRC-5 fibroblasts and exhibited significant anticancer potency (p < 0.001) against the H1437 (IC50 values: 62.50 μg/ml methanol extract and 125 μg/ml WE), H1573 (IC50 value: 125 μg/ml for both extracts) and A549 (IC50 value: 500 μg/ml ME). The water extract had no effect on the viability of A549 cells. Treated H1437 cells underwent p53-dependent apoptosis and S-phase cell cycle arrest while H1573 treated cells underwent p53-independed apoptosis and G0/G1 cell cycle arrest through upregulation of p21 mRNA expression levels. The expression levels of STAT1, STAT3, STAT5A and STAT5B genes increased significantly (p < 0.001) following the treatment of H1573 cells with ME and WE. Treatment of H1437 cells with ME upregulated the STAT1, STAT3, STAT5A and STAT5B mRNAs. Our results indicate that the proliferative inhibitory effect of D. calcarata extracts on A549 and H1573 cells is correlated with the suppression of Bcl-2, STAT3 and STAT5B while that is not the case in H1437 cells. Thus, our results suggest that the dysregulation of anti-apoptotic molecules Bcl-2, STAT3, STAT5A and STAT5B in H1437 may play a role in cancer cell survival, which may consequently contribute to the development of p53-mutated non-small human lung cancer. Our results indicate that D. calcarata is a promising source of anticancer agents for the treatment of p53-mutant human non-small lung cancer cells than the p53-wild type human non-small lung cancer cells.
Collapse
|
23
|
Ren C, Liu K, Zhao X, Guo H, Luo Y, Chang J, Gao X, Lv X, Zhi X, Wu X, Jiang H, Chen Q, Li Y. Research Progress of Traditional Chinese Medicine in Treatment of Myocardial fibrosis. Front Pharmacol 2022; 13:853289. [PMID: 35754495 PMCID: PMC9213783 DOI: 10.3389/fphar.2022.853289] [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: 01/12/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Effective drugs for the treatment of myocardial fibrosis (MF) are lacking. Traditional Chinese medicine (TCM) has garnered increasing attention in recent years for the prevention and treatment of myocardial fibrosis. This Article describes the pathogenesis of myocardial fibrosis from the modern medicine, along with the research progress. Reports suggest that Chinese medicine may play a role in ameliorating myocardial fibrosis through different regulatory mechanisms such as reduction of inflammatory reaction and oxidative stress, inhibition of cardiac fibroblast activation, reduction in extracellular matrix, renin-angiotensin-aldosterone system regulation, transforming growth Factor-β1 (TGF-β1) expression downregulation, TGF-β1/Smad signalling pathway regulation, and microRNA expression regulation. Therefore, traditional Chinese medicine serves as a valuable source of candidate drugs for exploration of the mechanism of occurrence and development, along with clinical prevention and treatment of MF.
Collapse
Affiliation(s)
- Chunzhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinke Zhao
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Huan Guo
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yali Luo
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Juan Chang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinfang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xiaodong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- The Second Hospital of Lanzhou University, Lanzhou, China
| | - Hugang Jiang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qilin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yingdong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
24
|
Shi CJ, Li SY, Shen CH, Pan FF, Deng LQ, Fu WM, Wang JY, Zhang JF. Icariside II suppressed tumorigenesis by epigenetically regulating the circβ-catenin-Wnt/β-catenin axis in colorectal cancer. Bioorg Chem 2022; 124:105800. [DOI: 10.1016/j.bioorg.2022.105800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
|
25
|
Deoxyelephantopin and Its Isomer Isodeoxyelephantopin: Anti-Cancer Natural Products with Multiple Modes of Action. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072086. [PMID: 35408483 PMCID: PMC9000713 DOI: 10.3390/molecules27072086] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 01/05/2023]
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. The development of cancer involves aberrations in multiple pathways, representing promising targets for anti-cancer drug discovery. Natural products are regarded as a rich source for developing anti-cancer therapies due to their unique structures and favorable pharmacology and toxicology profiles. Deoxyelephantopin and isodeoxyelephantopin, sesquiterpene lactone compounds, are major components of Elephantopus scaber and Elephantopus carolinianus, which have long been used as traditional medicines to treat multiple ailments, including liver diseases, diabetes, bronchitis, fever, diarrhea, dysentery, cancer, renal disorders, and inflammation-associated diseases. Recently, deoxyelephantopin and isodeoxyelephantopin have been extensively explored for their anti-cancer activities. This review summarizes and discusses the anti-cancer activities of deoxyelephantopin and isodeoxyelephantopin, with an emphasis on their modes of action and molecular targets. Both compounds disrupt several processes involved in cancer progression by targeting multiple signaling pathways deregulated in cancers, including cell cycle and proliferation, cell survival, autophagy, and invasion pathways. Future directions of research on these two compounds towards anti-cancer drug development are discussed.
Collapse
|
26
|
Aguchem RN, Okagu IU, Okagu OD, Ndefo JC, Udenigwe CC. A review on the techno‐functional, biological, and health‐promoting properties of hempseed‐derived proteins and peptides. J Food Biochem 2022; 46:e14127. [DOI: 10.1111/jfbc.14127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/25/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Rita Ngozi Aguchem
- Department of Biochemistry, Faculty of Biological Sciences University of Nigeria Nsukka Nigeria
| | | | - Ogadimma Desmond Okagu
- Department of Chemistry and Biomolecular Sciences, Faculty of Science University of Ottawa Ottawa Ontario Canada
| | - Joseph Chinedum Ndefo
- Department of Science Laboratory Technology, Faculty of Physical Sciences University of Nigeria Nsukka Nigeria
| | - Chibuike C. Udenigwe
- Department of Chemistry and Biomolecular Sciences, Faculty of Science University of Ottawa Ottawa Ontario Canada
- School of Nutrition Sciences, Faculty of Health Sciences University of Ottawa Ottawa Ontario Canada
| |
Collapse
|
27
|
Jayathilake AG, Kadife E, Kuol N, Luwor RB, Nurgali K, Su XQ. Krill oil supplementation reduces the growth of CT-26 orthotopic tumours in Balb/c mice. BMC Complement Med Ther 2022; 22:34. [PMID: 35120511 PMCID: PMC8817584 DOI: 10.1186/s12906-022-03521-4] [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: 08/06/2021] [Accepted: 01/25/2022] [Indexed: 12/09/2022] Open
Abstract
Background We have previously reported that the free fatty acid extract (FFAE) of krill oil (KO) significantly inhibits the proliferation and migration, and induces apoptosis of colorectal cancer (CRC) cells. This study aimed to investigate the in vivo efficacy of various doses of KO supplementation on the inhibition of CRC tumour growth, molecular markers of proliferation, angiogenesis, apoptosis, the epidermal growth factor receptor (EGFR) and its downstream molecular signalling. Methods Male Balb/c mice were randomly divided into four groups with five in each group. The control (untreated) group received standard chow diet; and other three groups received KO supplementation at 5%, 10%, and 15% of their daily dietary intake respectively for three weeks before and after the orthotopic implantation of CT-26 CRC cells in their caecum. The expression of cell proliferation marker Ki-67 and angiogenesis marker CD-31 were assessed by immunohistochemistry. The expression of EGFR, phosphorylated EGFR (pEGFR), protein kinase B (AKT), pAKT, extracellular signal-regulated kinase (ERK1/2), pERK1/2, cleaved caspase-7, cleaved poly (ADP-ribose) polymerase (PARP), and DNA/RNA damage were determined by western blot. Results KO supplementation reduced the CRC tumour growth in a dose-dependent manner; with 15% of KO being the most effective in reduction of tumour weight and volume (68.5% and 68.3% respectively, P < 0.001), inhibition of cell proliferation by 69.9% (P < 0.001) and microvessel density by 72.7% (P < 0.001). The suppressive effects of KO on EGFR and its downstream signalling, ERK1/2 and AKT, were consistent with our previous in vitro observations. Furthermore, KO exhibited pro-apoptotic effects on tumour cells as indicated by an increase in the expression of cleaved PARP by 3.9-fold and caspase-7 by 8.9-fold. Conclusions This study has demonstrated that KO supplementation reduces CRC tumour growth by inhibiting cancer cell proliferation and blood vessel formation and inducing apoptosis of tumour cells. These anti-cancer effects are associated with the downregulation of the EGFR signalling pathway and activation of caspase-7, PARP cleavage, and DNA/RNA damage. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03521-4.
Collapse
Affiliation(s)
| | - Elif Kadife
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia
| | - Nyanbol Kuol
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia
| | - Rodney Brain Luwor
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia.,Department of Medicine, Western Health, The University of Melbourne, Melbourne, Australia.,Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Sciences (AIMSS), Melbourne, Australia
| | - Xiao Qun Su
- Institute for Health and Sport, Victoria University, Melbourne, 8001, Australia.
| |
Collapse
|
28
|
Luo T, Jiang JG. Anticancer Effects and Molecular Target of Theaflavins from Black Tea Fermentation in Vitro and in Vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:15052-15065. [PMID: 34878780 DOI: 10.1021/acs.jafc.1c05313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Black tea is one of the most popular beverages in the world, and numerous epidemiological studies have shown that drinking black tea is good for health. As a natural tea pigment formed during the fermentation of black tea, the content of theaflavins accounts for only 2-6% of the dry weight of black tea, but they have a great impact on the color and taste of black tea soup. Recently, a large number of studies have shown that theaflavins have a significant anticancer effect. In this Perspective, we first state the physical and chemical properties, separation and purification methods, and biological formation pathways of theaflavins and analyze their safety and oral bioavailability and the structure-activity relationship of their antioxidant and anticancer activities; then, we describe in detail their anticancer effect in vitro and in vivo and highlight their various molecular targets involved in cancer inhibition. The anticancer molecular targets of theaflavins are mainly cell-cycle regulatory proteins, apoptosis-related proteins, cell-migration-related proteins, and growth transcription factors. Finally, the possibility of developing new health-care food based on theaflavins is discussed. This Perspective is expected to provide a theoretical basis for the anticancer application of theaflavins in the future.
Collapse
Affiliation(s)
- Ting Luo
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| | - Jian-Guo Jiang
- College of Food and Bioengineering, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
29
|
Winitchaikul T, Sawong S, Surangkul D, Srikummool M, Somran J, Pekthong D, Kamonlakorn K, Nangngam P, Parhira S, Srisawang P. Calotropis gigantea stem bark extract induced apoptosis related to ROS and ATP production in colon cancer cells. PLoS One 2021; 16:e0254392. [PMID: 34343190 PMCID: PMC8330925 DOI: 10.1371/journal.pone.0254392] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
Conventional chemotherapeutic agents for colorectal cancer (CRC) cause systemic side effects and eventually become less efficacious owing to the development of drug resistance in cancer cells. Therefore, new therapeutic regimens have focused on the use of natural products. The anticancer activity of several parts of Calotropis gigantea has been reported; however, the effects of its stem bark extract on inhibition of cancer cell proliferation have not yet been examined. In this study, the anticancer activity of C. gigantea stem bark extract, both alone and in combination with 5-fluorouracil (5-FU), was evaluated. A crude ethanolic extract was prepared from dry, powdered C. gigantea barks using 95% ethanol. This was then partitioned to obtain dichloromethane (CGDCM), ethyl acetate, and water fractions. Quantitative analysis of the constituent secondary metabolites and calotropin was performed. These fractions exhibited cytotoxicity in HCT116 and HT-29 cells, with CGDCM showing the highest potency in both the cell lines. A combination of CGDCM and 5-FU significantly enhanced the cytotoxic effect. Moreover, the resistance of normal fibroblast, HFF-1, cells to this combination demonstrated its safety in normal cells. The combination significantly enhanced apoptosis through the mitochondria-dependent pathway. Additionally, the combination reduced adenosine triphosphate production and increased the production of reactive oxygen species, demonstrating the mechanisms involved in the induction of apoptosis. Our results suggest that CGDCM is a promising anti-cancer agent and may enhance apoptosis induction by 5-FU in the treatment of CRC, while minimizing toxicity toward healthy cells.
Collapse
Affiliation(s)
- Thanwarat Winitchaikul
- Faculty of Medical Science, Department of Physiology, Naresuan University, Phitsanulok, Thailand
| | - Suphunwadee Sawong
- Faculty of Medical Science, Department of Physiology, Naresuan University, Phitsanulok, Thailand
| | - Damratsamon Surangkul
- Faculty of Medical Science, Department of Biochemistry, Naresuan University, Phitsanulok, Thailand
| | - Metawee Srikummool
- Faculty of Medical Science, Department of Biochemistry, Naresuan University, Phitsanulok, Thailand
| | - Julintorn Somran
- Faculty of Medicine, Department of Pathology, Naresuan University, Phitsanulok, Thailand
| | - Dumrongsak Pekthong
- Faculty of Pharmaceutical Sciences, Department of Pharmacy Practice, Naresuan University, Phitsanulok, Thailand
| | - Kittiya Kamonlakorn
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry and Pharmacognosy, Naresuan University, Phitsanulok, Thailand
| | - Pranee Nangngam
- Faculty of Science, Department of Biology, Naresuan University, Phitsanulok, Thailand
| | - Supawadee Parhira
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutical Technology, Naresuan University, Phitsanulok, Thailand
- * E-mail: (SP); (PS)
| | - Piyarat Srisawang
- Faculty of Medical Science, Department of Physiology, Naresuan University, Phitsanulok, Thailand
- * E-mail: (SP); (PS)
| |
Collapse
|
30
|
Rutin Mediated Apoptotic Cell Death in Caski Cervical Cancer Cells via Notch- 1 and Hes- 1 Downregulation. Life (Basel) 2021; 11:life11080761. [PMID: 34440505 PMCID: PMC8400226 DOI: 10.3390/life11080761] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Natural dietary molecules such as flavonoids have been recognized for their immense potential in cancer therapeutics with several health benefits. Hes-1 and Notch-1 overexpression has been associated with the progression of cervical cancer. However, the apoptosis-inducing potential of one such potent flavanol against these two key components of the Notch signaling pathway in cervical cancer has not been elucidated to date. Therefore, in this study, we performed several in vitro assays to gain detailed insight about the apoptotic inducing effect of rutin as well as its modulatory effect on Notch-1 and Hes-1 in cervical cancer cells. The results indicated that rutin led to a dose-dependent antiproliferative effects on Caski cervical cancer cells. DAPI and Mitotracker red staining revealed that rutin induced significant apoptotic effects via caspase-3/9 activation, ROS generation, and alteration in Bax/Bcl2 mRNA expression. Cell cycle analysis resulted in the arrest of cell cycle progression in G0/G1 that was associated with a reduced expression of CDK4 and Cyclin D1. The gene expression analysis further revealed that rutin treatment decreases Notch-1 and Hes-1 mRNA expression. Altogether, these results showed that rutin showed potent anticancer effects in human cervical cancer Caski cells by triggering apoptosis, G0/G1 phase arrest, and downregulating the level of Notch-1 and Hes-1 of the Notch signaling pathway.
Collapse
|
31
|
Anti-Proliferative and Anti-Migratory Activities of Hispidulin on Human Melanoma A2058 Cells. Biomolecules 2021; 11:biom11071039. [PMID: 34356663 PMCID: PMC8301921 DOI: 10.3390/biom11071039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 11/30/2022] Open
Abstract
Melanoma represents less than 5% of skin cancers, but is the most lethal, mainly because of its high-metastatic potential and resistance to various therapies. Therefore, it is important to develop effective treatments, especially chemotherapeutic drugs with cytotoxicity, anti-metastaticity, and few side effects. One such natural product is hispidulin, a flavone distributed in plants of the Asteraceae. Previous studies have demonstrated that hispidulin has various pharmacological benefits, such as anti-tumor, anti-inflammation, and anti-allergic effects. This study aims to explore the effects of hispidulin against melanoma in vitro and in vivo. Results revealed that hispidulin selectively decreased the cell viability of A2058 cells in a dose- and time-dependent manner. Hispidulin induced cells accumulated in the sub-G1 phase via activating caspase 8 and 9, increased cleaved caspase 3, and cleaved PARP expression. Hispidulin was able to decrease AKT and ERK phosphorylation, which facilitated cell growth and survival. Moreover, hispidulin promoted reactive oxygen species generation in cells and suppressed cell migration through downregulated matrix metalloproteinase-2 expression. Hispidulin significantly inhibited tumor growth in a xenograft model. Based on these results, hispidulin produces its anti-melanoma effects by inducing cancer cell apoptosis and reducing its migration. Therefore, we suggest hispidulin as a potent therapeutic candidate for melanoma treatment.
Collapse
|
32
|
Farghadani R, Naidu R. Curcumin: Modulator of Key Molecular Signaling Pathways in Hormone-Independent Breast Cancer. Cancers (Basel) 2021; 13:cancers13143427. [PMID: 34298639 PMCID: PMC8307022 DOI: 10.3390/cancers13143427] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer remains the most commonly diagnosed cancer and the leading cause of cancer death among females worldwide. It is a highly heterogeneous disease, classified according to hormone and growth factor receptor expression. Patients with triple negative breast cancer (TNBC) (estrogen receptor-negative/progesterone receptor-negative/human epidermal growth factor receptor (HER2)-negative) and hormone-independent HER2 overexpressing subtypes still represent highly aggressive behavior, metastasis, poor prognosis, and drug resistance. Thus, new alternative anticancer agents based on the use of natural products have been receiving enormous attention. In this regard, curcumin is a promising lead in cancer drug discovery due its ability to modulate a diverse range of molecular targets and signaling pathways. The current review has emphasized the underlying mechanism of curcumin anticancer action mediated through the modulation of PI3K/Akt/mTOR, JAK/STAT, MAPK, NF-ĸB, p53, Wnt/β-catenin, apoptosis, and cell cycle pathways in hormone-independent breast cancer, providing frameworks for future studies and insights to improve its efficiency in clinical practice. Abstract Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death among women worldwide. Despite the overall successes in breast cancer therapy, hormone-independent HER2 negative breast cancer, also known as triple negative breast cancer (TNBC), lacking estrogens and progesterone receptors and with an excessive expression of human epidermal growth factor receptor 2 (HER2), along with the hormone-independent HER2 positive subtype, still remain major challenges in breast cancer treatment. Due to their poor prognoses, aggressive phenotype, and highly metastasis features, new alternative therapies have become an urgent clinical need. One of the most noteworthy phytochemicals, curcumin, has attracted enormous attention as a promising drug candidate in breast cancer prevention and treatment due to its multi-targeting effect. Curcumin interrupts major stages of tumorigenesis including cell proliferation, survival, angiogenesis, and metastasis in hormone-independent breast cancer through the modulation of multiple signaling pathways. The current review has highlighted the anticancer activity of curcumin in hormone-independent breast cancer via focusing on its impact on key signaling pathways including the PI3K/Akt/mTOR pathway, JAK/STAT pathway, MAPK pathway, NF-ĸB pathway, p53 pathway, and Wnt/β-catenin, as well as apoptotic and cell cycle pathways. Besides, its therapeutic implications in clinical trials are here presented.
Collapse
|
33
|
Khalil AAK, Qazi AS, Nasir A, Ahn MJ, Shah MA, Ahmad MS, Sajjad W, Ali T, Naeem M, Shah FA, Khan MTA, Romman M, Shahfiq Ur Rehman, Haider A, Noor R. 2-Methoxy-6-Acetyl-7-Methyljuglone: A Bioactive Phytochemical with Potential Pharmacological Activities. Anticancer Agents Med Chem 2021; 22:687-693. [PMID: 34165415 DOI: 10.2174/1871520621666210623095636] [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: 12/20/2020] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 12/24/2022]
Abstract
Natural products have been the focus of biomedical and pharmaceutical research to develop new therapies in recent years. 2-methoxy-6-acetyl-7-methyljuglone (2-methoxystypandrone, MAM), a natural bioactive juglone derivative, is known to have various levels of pharmacotherapeutic efficacies as an anti-inflammatory, anticancer, antioxidant, antimicrobial, and anti-HIV activity. MAM fights cancer progression by inducing apoptosis, necroptosis, and deregulating signaling pathways through H2O2-induced JNK/iNOS/NO and MAPK, ERK1/2 pathways, JNK activation, and the RIP1/RIP3 complex. In this review, we summarize the pharmacological importance of MAM in the field of drug discovery. Furthermore, this review not only emphasizes the medicinal properties of MAM but also discusses its potential efficacy in future medicinal products.
Collapse
Affiliation(s)
- Atif Ali Khan Khalil
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Asma Saleem Qazi
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Abdul Nasir
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Mi-Jeong Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea
| | - Muhammad Ajmal Shah
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine; Hotchkiss Brain Institute, Cumming School of Medicine; University of Calgary, Alberta, T2N 4Z6. Canada
| | - Muhammad Saad Ahmad
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Wasim Sajjad
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Tahir Ali
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine; Hotchkiss Brain Institute, Cumming School of Medicine; University of Calgary, Alberta, T2N 4Z6. Canada
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Fawad Ali Shah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | | | - Muhammad Romman
- Department of Botany, University of Chitral, Chitral, Pakistan
| | - Shahfiq Ur Rehman
- Department of Rehabilitation, North West Institute of Health Sciences, Peshawar, Pakistan
| | - Adnan Haider
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Raishma Noor
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| |
Collapse
|
34
|
Jin JO, Chauhan PS, Arukha AP, Chavda V, Dubey A, Yadav D. The Therapeutic Potential of the Anticancer Activity of Fucoidan: Current Advances and Hurdles. Mar Drugs 2021; 19:265. [PMID: 34068561 PMCID: PMC8151601 DOI: 10.3390/md19050265] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
Several types of cancers share cellular and molecular behaviors. Although many chemotherapy drugs have been designed to weaken the defenses of cancer cells, these drugs may also have cytotoxic effects on healthy tissues. Fucoidan, a sulfated fucose-based polysaccharide from brown algae, has gained much attention as an antitumor drug owing to its anticancer effects against multiple cancer types. Among the anticancer mechanisms of fucoidan are cell cycle arrest, apoptosis evocation, and stimulation of cytotoxic natural killer cells and macrophages. Fucoidan also protects against toxicity associated with chemotherapeutic drugs and radiation-induced damage. The synergistic effect of fucoidan with existing anticancer drugs has prompted researchers to explore its therapeutic potential. This review compiles the mechanisms through which fucoidan slows tumor growth, kills cancer cells, and interacts with cancer chemotherapy drugs. The obstacles involved in developing fucoidan as an anticancer agent are also discussed in this review.
Collapse
Affiliation(s)
- Jun-O. Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Pallavi Singh Chauhan
- Amity Institute of Biotechnology, Amity University Madhya Pradesh, Gwalior 474005, India;
| | - Ananta Prasad Arukha
- Comparative Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA;
| | - Vishal Chavda
- Division of Anaesthesia, Sardar Women’s Hospital, Ahmedabad 380004, Gujarat, India;
| | - Anuj Dubey
- Department of Chemistry, ITM Group of Institutions, Gwalior 475005, India;
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
35
|
Islam Khan MZ, Law HKW. Cancer Susceptibility Candidate 9 (CASC9) Promotes Colorectal Cancer Carcinogenesis via mTOR-Dependent Autophagy and Epithelial-Mesenchymal Transition Pathways. Front Mol Biosci 2021; 8:627022. [PMID: 34017854 PMCID: PMC8129023 DOI: 10.3389/fmolb.2021.627022] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer worldwide. Many recent studies have demonstrated that different long non-coding RNAs (lncRNAs) are involved in the initiation, advancement, and metastasis of many cancers including CRC. Cancer susceptibility candidate 9 (CASC9) is an lncRNA that has been reported in many cancers, but its role in CRC is poorly understood. In this study, we aimed to examine the expression of CASC9 in CRC cell lines and to determine the mechanism of action of CASC9 in CRC carcinogenesis. METHODS The expression of CASC9 in CRC tissues was compared with normal samples from publicly available datasets in The Cancer Genome Atlas (TCGA) and The Encyclopedia of RNA Interactomes (ENCORI). CASC9 expression was further verified in four CRC cell lines (DLD1, HT-29, SW480, and HCT-116) and normal colorectal cell line (CCD-112CoN) by real-time quantitative polymerase chain reaction (RT-qPCR). After gene silencing in HCT-116 and SW480, Cell Counting Kit-8 assay, clonogenic assay, and wound healing assay were performed to evaluate cell proliferation, viability, and migration index of cells. Western blotting was used to explore the key pathways involved. RESULTS CASC9 was significantly upregulated as analyzed from both public datasets TCGA and ENCORI where its overexpression was associated with poor survival of CRC patients. Similarly, CASC9 was significantly overexpressed in the CRC cell lines compared with normal cells studied. The silencing of CASC9 in HCT-116 and SW480 attenuated cell proliferation and migration significantly. Furthermore, pathways investigations showed that silencing of CASC9 significantly induced autophagy, promoted AMP-activated protein kinase (AMPK) phosphorylation, inhibited mTOR and AKT signaling pathways, and altered epithelial-mesenchymal transition (EMT) marker protein expression. CONCLUSION We demonstrated that silencing of CASC9 contributes to the reduced CRC cell proliferation and migration by regulating autophagy and AKT/mTOR/EMT signaling. Therefore, CASC9 plays an important role in carcinogenesis, and its expression may act as a prognostic biomarker and a potential therapeutic target of CRC management.
Collapse
Affiliation(s)
| | - Helen Ka Wai Law
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
36
|
Adnan M, Rasul A, Hussain G, Shah MA, Sarfraz I, Nageen B, Riaz A, Khalid R, Asrar M, Selamoglu Z, Adem Ş, Sarker SD. Physcion and Physcion 8-O-β-D-glucopyranoside: Natural Anthraquinones with Potential Anticancer Activities. Curr Drug Targets 2021; 22:488-504. [PMID: 33050858 DOI: 10.2174/1389450121999201013154542] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/18/2020] [Accepted: 06/09/2020] [Indexed: 11/22/2022]
Abstract
Nature has provided prodigious reservoirs of pharmacologically active compounds for drug development since times. Physcion and physcion 8-O-β-D-glucopyranoside (PG) are bioactive natural anthraquinones which exert anti-inflammatory and anticancer properties with minimum or no adverse effects. Moreover, physcion also exhibits anti-microbial and hepatoprotective properties, while PG is known to have anti-sepsis as well as ameliorative activities against dementia. This review aims to highlight the natural sources and anticancer activities of physcion and PG, along with associated mechanisms of actions. On the basis of the literature, physcion and PG regulate multitudinous cell signaling pathways through the modulation of various regulators of cell cycle, protein kinases, microRNAs, transcriptional factors, and apoptosis linked proteins resulting in the effective killing of cancerous cells in vitro as well as in vivo. Both compounds effectively suppress metastasis, furthermore, physcion acts as an inhibitor of 6PGD and also plays an important role in chemosensitization. This review article suggests that physcion and PG are potent anticancer drug candidates, but further investigations on their mechanism of action and pre-clinical trials are mandatory in order to comprehend the full potential of these natural cancer killers in anticancer remedies.
Collapse
Affiliation(s)
- Muhammad Adnan
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Bushra Nageen
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Ammara Riaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Rida Khalid
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Muhammad Asrar
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Zeliha Selamoglu
- Department of Medical Biology, Faculty of Medicine, Nigde Ömer Halisdemir University, Nigde, Campus 51240, Turkey
| | - Şevki Adem
- Department of Chemistry, Faculty of Sciences, Cankiri Karatekin University, UluyazI Campus Cankiri, Turkey
| | - Satyajit D Sarker
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, England, United Kingdom
| |
Collapse
|
37
|
Wei L, Dong Y, Sun Y, Mei X, Ma X, Shi J, Yang Q, Ji Y, Zhang Z, Sun H, Sun X, Song S. Anticancer property of Hemp Bioactive Peptides in Hep3B liver cancer cells through Akt/GSK3β/β-catenin signaling pathway. Food Sci Nutr 2021; 9:1833-1841. [PMID: 33841802 PMCID: PMC8020916 DOI: 10.1002/fsn3.1976] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/04/2020] [Accepted: 10/10/2020] [Indexed: 12/11/2022] Open
Abstract
Foodborne protein hydrolysates exhibit biological activity that may be therapeutic in a number of human disease settings. Hemp peptides (HP) generated by controlled hydrolysis of hemp proteins have a number of health benefits and are of pharmaceutical value. In the present study, we produce small molecular weight HP from hemp seed and investigate its anticancer properties in Hep3B human liver cancer cells. We demonstrate that HP treatment increased apoptosis, reduced cell viability, and reduced cell migration in Hep3B human liver cancer cells without affecting the normal liver cell line L02. We correlate these phenotypes with increased cellular ROS levels, upregulation of cleaved caspase 3 and Bad, and downregulation of antiapoptotic Bcl-2. HP treatment led to increased Akt and GSK-3β phosphorylation, with subsequent downregulation of β-catenin, suggesting β-catenin signaling modulation as a critical mechanism by which HP exhibits anticancer properties. Our findings suggest HP are of potential therapeutic interest for liver cancer treatment.
Collapse
Affiliation(s)
- Lian‐Hui Wei
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Yan Dong
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Yu‐Feng Sun
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Xue‐Song Mei
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Xue‐Song Ma
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Jie Shi
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Qing‐li Yang
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Yan‐Ru Ji
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Zheng‐Hai Zhang
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| | - Hu‐Nan Sun
- College of Life Science and TechnologyHeilongjiang Bayi Agricultural UniversityDaqingChina
| | - Xing‐Rong Sun
- Daqing Branch of the Heilongjiang Academy of Agricultural SciencesDaqingChina
| | - Shu‐Min Song
- Daqing Branch of the Heilongjiang Academy of SciencesDaqingChina
| |
Collapse
|
38
|
Zafar E, Maqbool MF, Iqbal A, Maryam A, Shakir HA, Irfan M, Khan M, Li Y, Ma T. A comprehensive review on anticancer mechanism of bazedoxifene. Biotechnol Appl Biochem 2021; 69:767-782. [PMID: 33759222 DOI: 10.1002/bab.2150] [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: 12/17/2020] [Accepted: 03/08/2021] [Indexed: 12/24/2022]
Abstract
Cancer is counted as a second leading cause of death among nontransmissible diseases. Identification of novel anticancer drugs is therefore necessary for the effective treatment of cancer. Conventional drug discovery is time consuming and expensive process. Unlike conventional drug discovery, drug repositioning offers a novel strategy for urgent drug discovery since it is a cost-effective and faster process. Bazedoxifene (BZA) is a synthetic selective estrogen receptor modulator, approved by the United States Food and Drug Administration for the treatment of osteoporosis in postmenopausal women. BZA is now being studied for its anticancer activity in various cancers including breast cancer, liver cancer, pancreatic cancer, colon cancer, head and neck cancer, medulloblastoma, brain cancer, and gastrointestinal cancer. Studies have reported that BZA is effective in reducing cancer progression through multiple mechanisms. BZA could effectively inhibit STAT3, PI3K/AKT, and MAPK signaling pathways and induce apoptosis. In addition to its anticancer activity as monotherapy, BZA has been shown to enhance the chemotherapeutic efficacy of clinical drugs such as paclitaxel, cisplatin, palbociclib, and oxaliplatin in multiple neoplasms. This review mainly focused on the anticancer activity, cellular targets, and anticancer mechanism of BZA, which may help the further design and conduct of research and repositioning it for oncological clinic trials.
Collapse
Affiliation(s)
- Erum Zafar
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | | | - Asia Iqbal
- Department of Wild Life and Ecology, University of Veternary and Animal Sciences, Ravi Campus, Patoki, Pakistan
| | - Amara Maryam
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Hafiz Abdullah Shakir
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Muhammad Khan
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
39
|
Wen L, Zhou T, Jiang Y, Chang SK, Yang B. Prenylated flavonoids in foods and their applications on cancer prevention. Crit Rev Food Sci Nutr 2021; 62:5067-5080. [PMID: 33543993 DOI: 10.1080/10408398.2021.1881437] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Functional foods play an important role in health care and chronic diseases prevention, particularly cancer. Prenylated flavonoids are presented in many food resources. They are recognized as neutraceuticals due to their diverse health benefits. Up to now, more than 1000 prenylated flavonoids have been identified in plants. Their food resources are reviewed in this paper. Due to the good safety and cancer prevention effect of prenylated flavonoids, this paper reviews the cancer prevention activities and mechanisms reported in last decade. The structure-activity relationship is discussed. Due to the limited availability in nature, the heterologously biosynthetic technique of prenylated flavonoids is discussed in this review. Inclusion of dietary prenylated flavonoids into human diet is highly desirable. This paper combines the up-to-date information and give a clear image regarding prenylated flavonoids as neutraceuticals.
Collapse
Affiliation(s)
- Lingrong Wen
- Department of Horticulture, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Ting Zhou
- Department of Horticulture, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China.,Department of Horticulture, University of Chinese Academy of Sciences, Beijing, China
| | - Yueming Jiang
- Department of Horticulture, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China.,Core Botanical Gardens, Chinese Academy of Sciences, Guangzhou, China.,Department of Horticulture, University of Chinese Academy of Sciences, Beijing, China
| | - Sui Kiat Chang
- Department of Horticulture, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Bao Yang
- Department of Horticulture, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China.,Core Botanical Gardens, Chinese Academy of Sciences, Guangzhou, China.,Department of Horticulture, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Nwachukwu K, Rhoads E, Meek S, Bardi M. Back to nature: herbal treatment, environmental enrichment, and social play can protect against unpredictable chronic stress in Long-Evans rats (Rattus norvegicus). Psychopharmacology (Berl) 2021; 238:2999-3012. [PMID: 34333673 PMCID: PMC8325775 DOI: 10.1007/s00213-021-05917-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/30/2021] [Indexed: 11/03/2022]
Abstract
The importance of integrative biobehavioral responses to complex challenges cannot be overlooked. In this study, the synergetic effects of icariin (a flavonoid present in the plant Epimedium brevicornum), natural enrichment (NaEn), and play behavior were investigated. Rats (n = 60) were assigned to standard housing or NaEn; these two groups were subsequently divided into controls, rats receiving icariin treatments, and rats receiving icariin and allowed to play with an individual from another cage. All rats were exposed to unpredictable mild stressors for 4 weeks. At the end of the treatment, a Forced Swim Task (FST) was conducted to assess emotional regulation during an inescapable acute challenge. Biological samples were collected weekly and before and after the FST to monitor endocrine changes. Corticosterone (CORT), dehydroepiandrosterone (DHEA), and testosterone (T) were assayed. We found that icariin had a significant effect on DHEA/CORT ratios and T levels. NaEn also had a significant effect on both CORT and DHEA, but not on T levels. Play did not appear to be significantly related to the endocrine changes. The strongest positive effects on emotional resilience were observed in NaEn rats that also received icariin. Our results confirmed that using multiple channels to stimulate adaptive responses can be effective in increasing the ability of an organism to face uncertainty. Considering how quickly our life can change due to unpredictable events, our data is instrumental to a better comprehension of the many aspects of integrative biobehavioral responses.
Collapse
Affiliation(s)
- Kiana Nwachukwu
- grid.262455.20000 0001 2205 6070Department of Psychology and Neuroscience, Randolph-Macon College, Ashland, VA 23005 USA
| | - Elizabeth Rhoads
- grid.262455.20000 0001 2205 6070Department of Psychology and Neuroscience, Randolph-Macon College, Ashland, VA 23005 USA
| | - Sarah Meek
- grid.262455.20000 0001 2205 6070Department of Psychology and Neuroscience, Randolph-Macon College, Ashland, VA 23005 USA
| | - Massimo Bardi
- Department of Psychology and Neuroscience, Randolph-Macon College, Ashland, VA, 23005, USA. .,134D Copley Science Center, Randolph-Macon College, Ashland, VA, 23005, USA.
| |
Collapse
|
41
|
Zhao J, Zhou H, An Y, Shen K, Yu L. Biological effects of corosolic acid as an anti-inflammatory, anti-metabolic syndrome and anti-neoplasic natural compound. Oncol Lett 2020; 21:84. [PMID: 33363621 DOI: 10.3892/ol.2020.12345] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence has indicated that corosolic acid exerts anti-diabetic, anti-obesity, anti-inflammatory, anti-hyperlipidemic and anti-viral effects. More importantly, corosolic acid has recently attracted much attention due to its anticancer properties and innocuous effects on normal cells. Furthermore, the increasing proportion of obese and/or diabetic populations has led to an epidemic of non-alcoholic fatty liver disease (NAFLD), which frequently progresses to hepatocellular carcinoma (HCC). Evidence has indicated that NAFLD is closely associated with the development of HCC and comprises a high risk factor. The present review summarizes the anticancer effects of corosolic acid in vitro and in vivo, and its related molecular mechanisms. It also describes the inhibitory effects of corosolic acid on the progression of NAFLD and its associated molecular mechanisms, providing guidance for future research on corosolic acid in NAFLD-related HCC prevention and treatment. To the best of our knowledge, a review of corosolic acid as an anticancer agent has not yet been reported. Due to its multitargeted activity in cancer cells, corosolic acid exerts anticancer effects when administered alone, and acts synergistically when administered with chemotherapeutic drugs, even in drug-resistant cells. In addition, as a novel tool to treat metabolic syndromes, corosolic acid uses the same mechanism in its action against cancer as that used in the progression of NAFLD-related HCC. Therefore, corosolic acid has been suggested as an agent for the prevention and treatment of NAFLD-related HCC.
Collapse
Affiliation(s)
- Jinwei Zhao
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| | - Hong Zhou
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| | - Yanan An
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| | - Keshu Shen
- Department of Hepatobiliary Medicine of Jilin Hepatobiliary Hospital, Changchun, Jilin 130062, P.R. China
| | - Lu Yu
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| |
Collapse
|
42
|
Wang S, Wang N, Huang X, Yang B, Zheng Y, Zhang J, Wang X, Lin Y, Wang Z. Baohuoside i suppresses breast cancer metastasis by downregulating the tumor-associated macrophages/C-X-C motif chemokine ligand 1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153331. [PMID: 32911383 DOI: 10.1016/j.phymed.2020.153331] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/14/2020] [Accepted: 08/31/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Breast cancer is the most common malignancy in women and metastasis is the leading cause of breast cancer-related deaths. Our previous studies have shown that XIAOPI formula, a newly approved drug by the State Food and Drug Administration of China (SFDA), can dramatically inhibit breast cancer metastasis by modulating the tumor-associated macrophages/C-X-C motif chemokine ligand 1 (TAMs/CXCL1) pathway. However, the bioactive compound accounting for the anti-metastatic effect of XIAOPI formula remains unclear. PURPOSE This study was designed to separate the anti-metastatic bioactive compound from XIAOPI formula and to elucidate its action mechanisms. STUDY DESIGN/METHODS TAMs/CXCL1 promoter activity-guided fractionation and multiple chemical structure identification approaches were conducted to screen the bioactive compound from XIAOPI formula. Breast cancer cells and TAMs were co-cultured in vitro or co-injected in vivo to simulate their coexistence. Multiple molecular biology experiments, zebrafish breast cancer xenotransplantation model and mouse breast cancer xenografts were applied to validate the anti-metastatic activity of the screened compound. RESULTS Bioactivity-guided fractionation identified baohuoside I (BHS) as the key bioactive compound of XIAOPI formula in inhibiting TAMs/CXCL1 promoter activity. Functional studies revealed that BHS could significantly inhibit the migration and invasion as well as the expression of metastasis-related proteins in both human and mouse breast cancer cells, along with decreasing the proportion of breast cancer stem cells (CSCs). Furthermore, BHS could suppress the M2 phenotype polarization of TAMs and therefore attenuate their CXCL1 expression and secretion. Notably, mechanistic investigations validated TAMs/CXCL1 as the crucial target of BHS in suppressing breast cancer metastasis as exogenous addition of CXCL1 significantly abrogated the anti-metastatic effect of BHS on breast cancer cells. Moreover, BHS was highly safe in vivo as it exhibited no observable embryotoxicity or teratogenic effect on zebrafish embryos. More importantly, BHS remarkably suppressed breast cancer metastasis and TAMs/CXCL1 activity in both zebrafish breast cancer xenotransplantation model and mouse breast cancer xenografts. CONCLUSION This study not only provides novel insights into TAMs/CXCL1 as a reliable screening target for anti-metastatic drug discovery, but also suggests BHS as a promising candidate drug for metastatic breast cancer treatment.
Collapse
Affiliation(s)
- Shengqi Wang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Neng Wang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China; College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaowei Huang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China; College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bowen Yang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Yifeng Zheng
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Juping Zhang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Xuan Wang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Yi Lin
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Zhiyu Wang
- Integrative Research Laboratory of Breast Cancer, the Research Center for Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine & the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, Guangdong, China; College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
43
|
Li S, Zhan Y, Xie Y, Wang Y, Liu Y. The Impact of Icariside II on Human Prostate Cancer Cell Proliferation, Mobility, and Autophagy via PI3K-AKT-mTOR Signaling Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4169-4178. [PMID: 33116405 PMCID: PMC7549881 DOI: 10.2147/dddt.s268524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022]
Abstract
Introduction The flavonol glycoside icariside II (ICA II) has been shown to exhibit a range of anti-tumor properties. Herein, we evaluated the impact of ICA II on human prostate cancer cell proliferation, motility, and autophagy, and we further evaluated the molecular mechanisms underlying these effects. Methods We treated DU145 human prostate cancer cells with a range of ICA II doses and then assessed their proliferation via CCK-8 assay, while flow cytometry was used to monitor apoptosis and cell cycle progression. We further utilized wound healing and transwell assays to probe the impact of ICA II on migration and invasion, and assessed autophagy via laser confocal fluorescence microscopy. Western blotting was further utilized to measure LC3-II/I, Beclin-1, P70S6K, PI3K, AKT, mTOR, phospho-AKT, phospho-mTOR, and phospho-P70S6K levels, with qRT-PCR being used to evaluate the expression of specific genes at the mRNA level. Results We found that ICA II was capable of mediating the dose- and time-dependent suppression of DU145 cell proliferation, causing these cells to enter a state of cell cycle arrest and apoptosis. We further determined that ICA II treatment was associated with significant impairment of prostate cancer cell migration and invasion, whereas autophagy was enhanced in treated cells relative to untreated controls. Conclusion Our results indicate that ICA II treatment is capable of suppressing human prostate tumor cell proliferation and migration while enhancing autophagy via modulating the PI3K-AKT-mTOR signaling pathway. As such, ICA II may be an ideal candidate drug for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Shuang Li
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, People's Republic of China
| | - Yunlu Zhan
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, People's Republic of China
| | - Yingwei Xie
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, People's Republic of China
| | - Yonghui Wang
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, People's Republic of China
| | - Yuexin Liu
- Department of Urology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, People's Republic of China
| |
Collapse
|
44
|
Deoxyelephantopin, a novel naturally occurring phytochemical impairs growth, induces G2/M arrest, ROS-mediated apoptosis and modulates lncRNA expression against uterine leiomyoma. Biomed Pharmacother 2020; 131:110751. [PMID: 33152917 DOI: 10.1016/j.biopha.2020.110751] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022] Open
Abstract
Deoxyelephantopin (DOE), a phytochemical, extracted and purified from Elephantopus scaber, has been shown to exhibit antitumor activities. Objective of the present study was to investigate anti-tumor and apoptosis-inducing properties of DOE against uterine leiomyoma (UL) and to explore their molecular mechanisms. Primary cell cultures from fresh UL tissue were established and maintained up to 12 passages. The cells exhibited continuous proliferation with 24 -h doubling time until 12 passages and was then subjected to molecular characterization. The growth inhibitory effect of DOE on UL cells was confirmed by colony formation, cellular senescence, AO/PI and DAPI staining. Fluorescent-activated cell sorting (FACS) assay for apoptosis and cell cycle arrest analysis revealed that DOE significantly (p < 0.05) inhibited the UL cell proliferation via cell cycle arrest at G2/M phase and induced apoptosis via ROS production by lowering mitochondrial membrane potential. Subsequently, the DOE induced ROS was markedly attenuated by co-treatment of N-Acetyl-Cysteine (NAC). Our quantitative RT-PCR and western blot results showed up-regulation of Bax, Caspase-3 and down-regulation of Bcl2, P53, αSMA, COL4A2, VEGF, PCNA, Cyclin B1 and oncogenic lncRNAs (H19, HOTAIR, BANCR and ROR) in DOE treated UL cells which further strengthen our findings. In conclusion, DOE inhibits growth of UL cells via cell cycle arrest at G2/M phase, induces ROS-dependent caspase-3-mediated mitochondrial intrinsic apoptotic pathway and down-regulation of oncogenic lncRNA in UL cells. Our findings suggest that DOE deserves for further systematic investigation in the uterine leiomyoma animal model as a novel apoptosis inducer for potential applications in the prevention or treatment of uterine leiomyoma.
Collapse
|
45
|
Hu D, Gu Y, Wu D, Zhang J, Li Q, Luo J, Li S, Yuan Z, Zhu B. Icariside II protects cardiomyocytes from hypoxia‑induced injury by upregulating the miR‑7‑5p/BTG2 axis and activating the PI3K/Akt signaling pathway. Int J Mol Med 2020; 46:1453-1465. [PMID: 32945347 PMCID: PMC7447325 DOI: 10.3892/ijmm.2020.4677] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
Icariside II (ICS II) has been reported to have protective effects against oxidative stress. However, whether ICS II protects cardiomyocytes from myocardial infarction (MI), and the associated underlying mechanisms, remain to be elucidated. Therefore, the current study investigated the effects of ICS II on hypoxia‑injured H9c2 cells, as well as the associated molecular mechanisms. A hypoxic injury model was established to emulate the effects of MI. The effects of ICS II on the proliferation of rat cardiomyocyte H9c2 cells were assessed with cell counting kit‑8 assays. The apoptotic status of the cells was assessed by flow cytometry, and the expression of apoptosis‑related proteins was analyzed by western blotting. A microRNA (miRNA/miR) microarray was used to quantify the differential expression of miRNAs after ICS II treatment, and the levels of miR‑7‑5p were further quantified by reverse transcription‑quantitative PCR. Whether ICS II affected hypoxia‑injured cells via miR‑7‑5p was subsequently examined, and the target of miR‑7‑5p was also investigated by bioinformatics analysis and luciferase reporter assays. The effects of ICS II on the PI3K/Akt pathway were then evaluated by western blot analysis. Hypoxia treatment decreased viability and the migration and invasion abilities of H9c2 cells, and also induced apoptosis. ICS II significantly increased viability and reduced hypoxia‑associated apoptosis. Moreover, ICS II treatment led to the upregulation of miR‑7‑5p, and the protective effects of ICS II were found to rely on miR‑7‑5p. Moreover, BTG anti‑proliferation factor (BTG2) was identified as a direct target of miR‑7‑5p, and overexpression of BTG2 inhibited the protective effects of miR‑7‑5p. Finally, ICS II treatment resulted in the activation of the PI3K/Akt signaling pathway, which is essential for the survival of H9c2 cells under hypoxic conditions. In summary, ICS II reduces hypoxic injury in H9c2 cells via the miR‑7‑5p/BTG2 axis and activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Dongxia Hu
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| | - Yong Gu
- Department of Clinical Laboratory, Jiangxi Thoracic Hospital, Nanchang, Jiangxi 330321, P.R. China
| | - Dan Wu
- Department of Neurology, Nanchang Hongdu Hospital of TCM, Nanchang, Jiangxi 330321, P.R. China
| | - Juanjuan Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| | - Qing Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| | - Jun Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| | - Shaochuan Li
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| | - Zhen Yuan
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| | - Bo Zhu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330321, P.R. China
| |
Collapse
|
46
|
Ashaq A, Maqbool MF, Maryam A, Khan M, Shakir HA, Irfan M, Qazi JI, Li Y, Ma T. Hispidulin: A novel natural compound with therapeutic potential against human cancers. Phytother Res 2020; 35:771-789. [PMID: 32945582 DOI: 10.1002/ptr.6862] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/29/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most devastating disease and leading cause of death worldwide. The conventional anticancer drugs are monotarget, toxic, expensive and suffer from drug resistance. Development of multi-targeted drugs from natural products has emerged as a new paradigm to overcome aforementioned conventionally encountered obstacles. Hispidulin (HIS), is a biologically active natural flavone with versatile biological and pharmacological activities. The anticancer, antimutagenic, antioxidative and anti-inflammatory properties of HIS have been reported. The aim of this review is to summarize the findings of several studies over the last few decades on the anticancer activity of HIS published in various databases including PubMed, Google Scholar, and Scopus. HIS has been shown to reduce the growth of cancer cells by inducing apoptosis, arresting cell cycle, inhibiting angiogenesis, invasion and metastasis via modulating multiple signaling pathways implicated in cancer initiation and progression. Multitargeted anticancer activity of HIS remains the strongest point for developing it into potential anticancer drug. We also highlighted the natural sources, anticancer mechanism, cellular targets, and chemo-sensitizing potential of HIS. This review will provide bases for design and conduct of further pre-clinical and clinical trials to develop HIS into a lead structure for future anticancer therapy.
Collapse
Affiliation(s)
- Aisha Ashaq
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | | | - Amara Maryam
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Khan
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Hafiz A Shakir
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Javed I Qazi
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
47
|
Kanwal N, Rasul A, Hussain G, Anwar H, Shah MA, Sarfraz I, Riaz A, Batool R, Shahbaz M, Hussain A, Selamoglu Z. Oleandrin: A bioactive phytochemical and potential cancer killer via multiple cellular signaling pathways. Food Chem Toxicol 2020; 143:111570. [PMID: 32640345 DOI: 10.1016/j.fct.2020.111570] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/17/2022]
Abstract
Nerium oleander, a member of family Apocynaceae, is commonly known as Kaner in various countries of Asia and Mediterranean region. This plant has been renowned to possess significant therapeutic potential due to its various bioactive compounds which have been isolated from this plant e.g., cardiac glycosides, oleandrin, α-tocopherol, digitoxingenin, urosolic acid, quercetin, odorosides, and adigoside. Oleandrin, a saponin glycoside is one of the most potent and pharmacologically active phytochemicals of N. oleander. Its remarkable pharmacotherapeutic potential have been interpreted as anticancer, anti-inflammatory, anti-HIV, neuroprotective, antimicrobial and antioxidant. This particular bioactive entity is known to target the multiple deregulated signaling cascades of cancer such as NF-κB, MAPK, and PI3K/Akt. The main focus of the current study is to comprehend the action mechanisms of oleandrin against various pathological conditions. The current review is a comprehensive summary to facilitate the researchers to understand the pharmacological position of the oleandrin in the arena of drug discovery, representing this compound as a new drug candidate for further researches. Moreover, in vivo and in silico based studies are required to explore the mechanistic approaches regarding the pharmacokinetics and biosafety profiling of this compound to completely track its candidature status in natural drug discovery.
Collapse
Affiliation(s)
- Nazia Kanwal
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan.
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, 38000, Pakistan.
| | - Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Ammara Riaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Rabia Batool
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Muhammad Shahbaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Arif Hussain
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Zeliha Selamoglu
- Department of Medical Biology, Faculty of Medicine, Nigde Omer Halisdemir University, Nidge Campus, 51240, Turkey
| |
Collapse
|
48
|
Khan M, Maryam A, Saleem MZ, Shakir HA, Qazi JI, Li Y, Ma T. Brevilin A induces ROS-dependent apoptosis and suppresses STAT3 activation by direct binding in human lung cancer cells. J Cancer 2020; 11:3725-3735. [PMID: 32328177 PMCID: PMC7171504 DOI: 10.7150/jca.40983] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022] Open
Abstract
Sesquiterpene lactones have been shown to be promising leads for anticancer drug development. Brevilin A (BLN-A), a sesquiterpene lactone compound of Centipeda minima has been shown to exhibit anticancer effects against various cancer cells. However, the anticancer mechanism and cellular targets of BLN-A remain elusive. Here in this study, BLN-A inhibits proliferation and induces cell morphological changes in A549 and NCI-H1650 non-small cell lung cancer cells in a dose-dependent manner. Moreover, BLN-A increased ROS generation and bax/bcl-2 ratio while decreased intracellular glutathione (GSH), and mitochondrial membrane potential which resulted in induction of apoptosis as evident by annexin-V/FITC staining, caspase-3 activation and PARP cleavage. Supplementation of cells with NAC (ROS Scavenger) effectively protected the cells from BLN-A-induced apoptosis. Finally, BLN-A inhibited constitutive as well as IL-6- and EGF-induced STAT3 activation at Tyr705. Using molecular docking and SPR analyses, we found that BLN-A directly binds with STAT3 and thereby inhibits its activation. Knocking down of STAT3 by stable transfection with shRNA suppressed growth and augmented cytotoxicity of BLN-A, indicating the key role of STAT3 in BLN-A-mediated apoptosis. Cumulative findings suggest that BLN-A is a promising lead structure for developing it into a potent STAT3 inhibitor and therapeutic agent against NSCLC as well.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | - Amara Maryam
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | | | - Hafiz Abdullah Shakir
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | - Javed Iqbal Qazi
- Department of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore-54590, Pakistan
| | - Yongming Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Tonghui Ma
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| |
Collapse
|
49
|
Huang C, Lu HF, Chen YH, Chen JC, Chou WH, Huang HC. Curcumin, demethoxycurcumin, and bisdemethoxycurcumin induced caspase-dependent and -independent apoptosis via Smad or Akt signaling pathways in HOS cells. BMC Complement Med Ther 2020; 20:68. [PMID: 32126993 PMCID: PMC7076840 DOI: 10.1186/s12906-020-2857-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background Osteosarcoma is the most common primary malignant bone tumor in children and adolescents and has also been associated with a high degree of malignancy and enhanced metastatic capacity. Curcumin (CUR) is well known for its anti-osteosarcoma activity. However, both demethoxycurcumin (DMC), and bisdemethoxycurcumin (BDMC) are natural curcumin analogues/congeners from turmeric whose role in osteosarcoma development remains unknown. Methods To evaluate the growth inhibitory effects of CUR, DMC and BDMC on osteosarcoma (HOS and U2OS), breast (MDA-MB-231), and melanoma (A2058) cancer cells, we employed the MTT assay, annexin V-FITC /7-AAD staining, and clonogenic assay. Results CUR,DMC, and BDMC all decreased the viability of HOS, U2OS, MDA-MB-231, and A2058 cancer cells. Additionally, CUR,DMC, and BDMC induced the apoptosis of HOS cells through activation of Smad 2/3 or repression of Akt signaling pathway. Furthermore, the combination of CUR,DMC, and BDMC synergistically reduced cell viability, colony formation and increased apoptosis than either two or a single agent in HOS cells. Conclusions The combination of these three compounds could be used as a novel target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Cheng Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, 11221, Taiwan.,Department of Earth and Life Sciences, University of Taipei, Taipei, 11153, Taiwan
| | - Hsu-Feng Lu
- Departments of Clinical Pathology, Cheng Hsin General Hospital, Taipei, 11221, Taiwan.,Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei, 24205, Taiwan
| | - Yu-Hsuan Chen
- Department of Applied Science, National Tsing Hua University South Campus, No.521, Nanda Rd, Hsinchu City, 30014, Taiwan
| | - Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, 60004, Taiwan
| | - Wen-Hsiang Chou
- School of Medicine, National Defense Medical Center, Taipei, 11490, Taiwan.,Department of Orthopedics, Cheng Hsin General Hospital, Taipei, 11220, Taiwan
| | - Hsiu-Chen Huang
- Department of Applied Science, National Tsing Hua University South Campus, No.521, Nanda Rd, Hsinchu City, 30014, Taiwan.
| |
Collapse
|
50
|
Liu Y, Yang H, Guo Q, Liu T, Jiang Y, Zhao M, Zeng K, Tu P. Cucurbitacin E Inhibits Huh7 Hepatoma Carcinoma Cell Proliferation and Metastasis via Suppressing MAPKs and JAK/STAT3 Pathways. Molecules 2020; 25:molecules25030560. [PMID: 32012950 PMCID: PMC7037518 DOI: 10.3390/molecules25030560] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/16/2020] [Accepted: 01/25/2020] [Indexed: 12/12/2022] Open
Abstract
Cucurbitacin E (CuE), a highly oxygenated tetracyclic triterpene from Cucurbitaceae, has shown to exhibit potent cytotoxic and anti-proliferative properties against several human cancer cells. However, the underlying effects and mechanisms of CuE regarding hepatocellular carcinoma (HCC) have not been well understood. In the current study, unbiased RNA-sequencing (RNA-seq) and bioinformatics analysis was applied to elucidate the underlying molecular mechanism. CuE could significantly inhibit cell proliferation and migration of Huh7 cells, meanwhile CuE exhibited potent anti-angiogenic activity. RNA-seq analysis revealed that CuE negatively regulated 241 differentially expressed genes (DEGs) involved in multiple processes including cytoskeleton formation, angiogenesis and focal adhesion. Further analysis revealed that CuE effectually regulated diversified pharmacological signaling pathways such as MAPKs and JAK-STAT3. Our findings demonstrated the role of CuE in inhibiting proliferation and migration, providing an insight into the regulation of multiple signaling pathways as a new paradigm for anti-cancer treatment strategy.
Collapse
|