1
|
Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural Products as Anticancer Agents: Current Status and Future Perspectives. Molecules 2022; 27:molecules27238367. [PMID: 36500466 PMCID: PMC9737905 DOI: 10.3390/molecules27238367] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Natural products have been an invaluable and useful source of anticancer agents over the years. Several compounds have been synthesized from natural products by modifying their structures or by using naturally occurring compounds as building blocks in the synthesis of these compounds for various purposes in different fields, such as biology, medicine, and engineering. Multiple modern and costly treatments have been applied to combat cancer and limit its lethality, but the results are not significantly refreshing. Natural products, which are a significant source of new therapeutic drugs, are currently being investigated as potential cytotoxic agents and have shown a positive trend in preclinical research and have prompted numerous innovative strategies in order to combat cancer and expedite the clinical research. Natural products are becoming increasingly important for drug discovery due to their high molecular diversity and novel biofunctionality. Furthermore, natural products can provide superior efficacy and safety due to their unique molecular properties. The objective of the current review is to provide an overview of the emergence of natural products for the treatment and prevention of cancer, such as chemosensitizers, immunotherapeutics, combinatorial therapies with other anticancer drugs, novel formulations of natural products, and the molecular mechanisms underlying their anticancer properties.
Collapse
Affiliation(s)
- Abid Naeem
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Zhang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yali Liu
- Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang Medical College, Nanchang 330006, China
- Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang Medical College, Nanchang 330006, China
| | - Weifeng Zhu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Correspondence:
| |
Collapse
|
2
|
Yin WM, Cao XB, Li SX, Zhang F, Guan YF. Brassinin inhibits proliferation and induces cell cycle arrest and apoptosis in nasopharyngeal cancer C666-1 cells. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
3
|
Song X, Greiner-Tollersrud OK, Zhou H. Oral Microbiota Variation: A Risk Factor for Development and Poor Prognosis of Esophageal Cancer. Dig Dis Sci 2022; 67:3543-3556. [PMID: 34505256 DOI: 10.1007/s10620-021-07245-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022]
Abstract
Recent studies have shown that oral microbiota play an important role in the esophageal cancer (EC) initiation and progression, suggesting that oral microbiota is a new risk factor for EC. The composition of the microbes inhabiting the oral cavity could be perturbed with continuous factors such as smoking, alcohol consumption, and inflammation. The microbial alteration involves the decrease of beneficial species and the increase of pathogenic species. Experimental evidences suggest a significant role of oral commensal organisms in protecting hosts against EC. By contrast, oral pathogens, especially Porphyromonas gingivalis and Fusobacterium nucleatum, give rise to the risk for developing EC through their pro-inflammatory and pro-tumorigenic activities. The presences of oral dysbiosis, microbial biofilm, and periodontitis in EC patients are found to be associated with invasive cancer phenotypes and poor prognosis. The mechanism of oral bacteria in EC progression is complex, which involves a combination of cytokines, chemokines, oncogenic signaling pathways, cell surface receptors, the degradation of extracellular matrix, and cell apoptosis. From a clinical perspective, good oral hygiene, professional oral care, and rational use of antibiotics bring positive impacts on oral microbial balance, thus helping individuals reduce the risk of EC, inhibiting postoperative complications among EC patients, and improving the efficiency of chemoradiotherapy. However, current oral hygiene practices mainly focus on the oral bacteria-based predictive and preventive purposes. It is still far from implementing microbiota-dependent regulation as a therapy for EC. Further explorations are needed to render oral microbiota a potential target for treating EC.
Collapse
Affiliation(s)
- Xiaobo Song
- Department of Microbiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, China.,Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, 9037, Tromsø, Norway
| | - Ole K Greiner-Tollersrud
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, 9037, Tromsø, Norway
| | - Huimin Zhou
- Department of Microbiology, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, Liaoning Province, China.
| |
Collapse
|
4
|
Zhou W, Yan X, Zhai Y, Liu H, Guan L, Qiao Y, Jiang J, Peng L. Phillygenin ameliorates nonalcoholic fatty liver disease via TFEB-mediated lysosome biogenesis and lipophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154235. [PMID: 35716542 DOI: 10.1016/j.phymed.2022.154235] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/21/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Lipophagy is an autophagic process, which delivers the intracellular lipid droplets to the lysosomes for degradation. Recent studies revealed that the impairment of lysosomal biogenesis and autophagic flux led to dysregulation of lipophagy in hepatocytes, which exacerbated the development of nonalcoholic fatty liver disease (NAFLD). Therefore, agents restoring autophagic flux and lipophagy in hepatocytes may have therapeutic potential against this increasingly prevalent disease. Phillygenin (PHI), a lignin extracted from Forsythia suspense, exerts hepatoprotective and anti-inflammatory effects. However, the effect of PHI on NAFLD remains unknown. PURPOSE This study aimed to investigate the protective effect of PHI on NAFLD and elucidate the underlying mechanism. METHODS The effects of PHI were examined in palmitate (PA)-stimulated AML12 cells and primary hepatocytes, as well as in NAFLD mice induced by a high-fat diet (HFD). We also used transcription factor EB (TFEB) knockdown hepatocytes and hepatocyte-specific TFEB knockout (TFEBΔhep) mice for mechanistic studies. In vivo and in vitro studies were performed using western blots, immunofluorescence techniques, and transmission electron microscopy. RESULTS Our results indicated that autophagic flux and lysosome biogenesis in PA-stimulated hepatocytes were impaired. PHI alleviated lipid deposition by increasing lysosomal biogenesis and autophagic flux. It also stimulated the release of endoplasmic reticulum Ca2+ to activate calcineurin, which regulated TFEB dephosphorylation and nuclear translocation, and promoted lysosomal biogenesis. In addition, PHI blocked the NLRP3 inflammasome pathway and improved hepatocyte inflammation in an autophagy-dependent manner. Consistent with the in vitro results, PHI improved hepatic steatosis and inflammation in HFD mice, but these beneficial effects were eliminated in hepatocyte-specific TFEB knockout mice. CONCLUSION Despite PHI has been reported to have anti-hepatic fibrosis effects, whether it has a hepatoprotective effects against NAFLD and the underlying molecular mechanism remain unclear. Herein, we found that PHI restored lipophagy and suppressed lipid accumulation and inflammation by regulating the Ca2+-calcineurin-TFEB axis in hepatocytes. Thus, PHI represents a therapeutic candidate for the treatment of NAFLD.
Collapse
Affiliation(s)
- Wenling Zhou
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China; College of Life Sciences, Hebei University, Baoding 071002, China
| | - Xu Yan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China; College of Life Sciences, Hebei University, Baoding 071002, China
| | - Yuanyuan Zhai
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China; College of Life Sciences, Hebei University, Baoding 071002, China
| | - Hao Liu
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Lingling Guan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yuan Qiao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jizhi Jiang
- College of Life Sciences, Hebei University, Baoding 071002, China.
| | - Liang Peng
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
5
|
Qu Q, Li Y, Dong Q, Li S, Du H, Wang Z, Gong X, Zhang W, Lv W, Chao L, Liu M, Tang X, Guo S. Comparative Evaluation of Forsythiae Fructus From Different Harvest Seasons and Regions by HPLC/NIR Analysis and Anti-inflammatory and Antioxidant Assays. Front Pharmacol 2021; 12:737576. [PMID: 34899295 PMCID: PMC8652199 DOI: 10.3389/fphar.2021.737576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
Forsythiae Fructus (FF), the dry fruit of Forsythia suspensa (Thunb.) Vahl, has a long history of use in traditional Chinese Medicine for its heat-clearing and detoxifying properties. It possesses clinical therapeutic effects and biological functions showing efficacy in handling different diseases. To investigate the FF differences in Henan, Shanxi, and Shaanxi in August and October, the surface morphology, mid-infrared and near-infrared spectrums, and HPLC were analyzed. Concurrently, the anti-inflammatory and antioxidant effects on LPS-induced J774A.1 cells were evaluated by western blot and RT-qPCR. The results showed that FF from different Harvest Seasons and Regions are provided with different microstructures and mid-infrared and near-infrared spectrums, and the levels of forsythiaside A and phillyrin of FF from Shanxi in August and phillygenin of FF from Shaanxi in August were the highest. Meanwhile, FF from Shanxi and Shaanxi in August markedly reduced the levels of inflammatory cytokines and mediators (TNF-α, IL-1β, NF-κB, and iNOS) and the protein expression levels of phosphorylated total IKKα/β and nuclear NF-κB. In August, SXFF and SAXFF also promoted the mRNA expression levels of HO-1 and NQO1 and the protein expression levels of HO-1 and nuclear Nrf2 and suppressed the protein expression levels of KEAP1. Spearman correlation analysis showed that phillygenin had a strong correlation with the protein expression on LPS-induced J774A.1 cells. In summary, our results showed that FF from harvest seasons and regions contributed to the distinct differences in microstructure, the mid-infrared and near-infrared spectrums, and compound content. More importantly, FF from Shanxi and Shaanxi in August showed marked anti-inflammatory and antioxidant activities, but with some differences, which may be because of different contents of phillygenin and phillyrin of lignans in FF.
Collapse
Affiliation(s)
- Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuefei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qi Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shupeng Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongliang Du
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhihua Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaopei Gong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenchang Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Weijie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Limin Chao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mengjie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinggang Tang
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Guangdong Research Center for Veterinary Traditional Chinese Medicine and Natural Medicine Engineering Technology, Guangzhou, China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
6
|
Li B, Yang H, Shen B, Huang J, Qin Z. Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 increases cellular proliferation and colony formation capacity in lung cancer via activation of E2F transcription factor 1. Oncol Lett 2021; 22:851. [PMID: 34733369 PMCID: PMC8561618 DOI: 10.3892/ol.2021.13112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 05/25/2021] [Indexed: 01/30/2023] Open
Abstract
Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 (PLOD1) is an enzyme that catalyzes the hydroxylation of lysyl residues in collagen-like peptides, and is responsible for the stability of intermolecular crosslinks. High PLOD1 mRNA levels have been determined to be prognostically significant in numerous human malignancies. The objective of the present study was to elucidate the pathological mechanism of PLOD1 in lung cancer. The expression status and prognostic value of PLOD1 in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSA) were investigated using Gene Expression Profiling Interactive Analysis (GEPIA). Cell Counting Kit 8 and colony formation assays were performed to assess the impact of PLOD1 depletion and overexpression on the proliferation and colony formation abilities of the A549 lung cancer cell line. Luciferase reporter assays were used to clarify whether E2F transcription factor 1 (E2F1) was a downstream target of PLOD1 in lung cancer. Finally, the correlations between PLOD1 expression and a typical central downstream effector molecule of E2F1 signaling were determined using cBioportal. The GEPIA datasets revealed that PLOD1 mRNA levels were upregulated in LUAD and LUSC samples. Furthermore, the overexpression of PLOD1 promoted cancer cell proliferation and colony formation in vitro, while PLOD1-knockout produced the opposite effect. Notably, PLOD1 markedly induced the transcriptional activity of E2F1. Additionally, the expression of PLOD1 was significantly correlated with that of H2A histone family member X. In conclusion, the findings of the present study indicate that PLOD1 promoted lung cancer through E2F1 activation, and proposed a rationale for targeting the PLOD1/E2F1 axis to treat lung cancer.
Collapse
Affiliation(s)
- Baijun Li
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Hao Yang
- Department of Administration, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Bin Shen
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Jianwei Huang
- Department of Thoracic Cardiovascular Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Zhiqiang Qin
- Department of Pulmonary and Critical Care Medicine, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
7
|
Ebrahimi M, Norouzi P, Aazami H, Moosavi-Movahedi AA. Review on oxidative stress relation on COVID-19: Biomolecular and bioanalytical approach. Int J Biol Macromol 2021; 189:802-818. [PMID: 34418419 PMCID: PMC8372478 DOI: 10.1016/j.ijbiomac.2021.08.095] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
COVID-19 disease has put life of people in stress worldwide from many aspects. Since the virus has mutated in absolutely short period of time the challenge to find a suitable vaccine has become harder. Infection to COVID-19, especially at severe life threatening states is highly dependent on the strength of the host immune system. This system is partially dependent on the balance between oxidative stress and antioxidant. Besides, this virus still has unknown mechanism of action companied by a probable commune period. From another hand, some reactive oxygen species (ROS) levels can be helpful on the state determination of the disease. Thus it could be possible to use modern bioanalytical techniques for their detection and determination, which could indicate the disease state at the golden time window since they have the potential to show whether specific DNA, RNA, enzymes and proteins are affected. This also could be used as a preclude study or a reliable pathway to define the best optimized time of cure beside effective medical actions. Herein, some ROS and their relation with SARS-CoV-2 virus have been considered. In addition, modern bioelectroanalytical techniques on this approach from quantitative and qualitative points of view have been reviewed.
Collapse
Affiliation(s)
- Mehrnaz Ebrahimi
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| | - Parviz Norouzi
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran, Iran.
| | - Hossein Aazami
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
8
|
Lin Y, Yang P. Phillygenin inhibits the inflammation and apoptosis of pulmonary epithelial cells by activating PPARγ signaling via downregulation of MMP8. Mol Med Rep 2021; 24:775. [PMID: 34490481 PMCID: PMC8441984 DOI: 10.3892/mmr.2021.12415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/10/2021] [Indexed: 01/01/2023] Open
Abstract
Acute lung injury (ALI) is often responsible for the high morbidity of critically ill patients. The present study aimed to investigate whether phillygenin (PHI) can inhibit inflammation and apoptosis of pulmonary epithelial cells by activating peroxisome proliferator-activated receptor γ (PPARγ) signaling. The in vitro model of ALI was established using lipopolysaccharide (LPS) and PHI was used to treat the LPS-induced cells. Cell viability was assessed using the MTT assay and the concentration levels of the inflammatory factors were detected by ELISA. Western blotting and reverse transcription-quantitative PCR were conducted to measure the expression levels of the inflammation- and apoptosis-associated proteins. The MMP8-overexpression plasmid was transfected into LPS-induced cells, which were treated with PHI treatment and the expression levels of PPARγ were detected via western blotting. PHI treatment suppressed the induction of inflammation and apoptosis of LPS-induced BEAS-2B cells. Furthermore, the expression levels of MMP8 in BEAS-2B cells induced by LPS were decreased following PHI treatment. Following transfection of the MMP8 overexpression plasmid into the LPS-induced BEAS-2B cells and subsequent treatment of these cells with PHI, the expression levels of PPARγ were decreased. In conclusion, it was shown that PHI inhibited the inflammation and apoptosis of pulmonary epithelial cells by activating PPARγ signaling via downregulating MMP8. These data may provide valuable information for future studies exploring the therapeutic effects of PHI for ALI.
Collapse
Affiliation(s)
- Yufeng Lin
- Department of Pediatrics, Gaolangang Hospital of Zhuhai People's Hospital, Zhuhai, Guangdong 519050, P.R. China
| | - Peng Yang
- Department of PICU, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
9
|
Ding X, Lu D, Fan J. A natural product phillygenin suppresses osteosarcoma growth and metastasis by regulating the SHP-1/JAK2/STAT3 signaling. Biosci Biotechnol Biochem 2021; 85:307-314. [PMID: 33604629 DOI: 10.1093/bbb/zbaa007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022]
Abstract
Osteosarcoma represents one of the most devastating cancers due to its high metastatic potency and fatality. Osteosarcoma is insensitive to traditional chemotherapy. Identification of a small molecule that blocks osteosarcoma progression has been a challenge in drug development. Phillygenin, a plant-derived tetrahydrofurofuran lignin, has shown to suppress cancer cell growth and inflammatory response. However, how phillygenin plays functional roles in osteosarcoma has remained unveiled. In this study, we showed that phillygenin inhibited osteosarcoma cell growth and motility in vitro. Further mechanistic studies indicated that phillygenin blocked STAT3 signaling pathway. Phillygenin led to significant downregulation of Janus kinase 2 and upregulation of Src homology region 2 domain-containing phosphatase 1. Gene products of STAT3 regulating cell survival and invasion were also inhibited by phillygenin. Therefore, our studies provided the first evidence that phillygenin repressed osteosarcoma progression by interfering STAT3 signaling pathway. Phillygenin is a potential candidate in osteosarcoma therapy.
Collapse
Affiliation(s)
- Xiaomin Ding
- Department of Orthopedics, Affiliated Hospital 2 of Nantong University, Jiangsu, China
| | - Danqing Lu
- Department of Orthopedics, The Second People's Hospital of Kunshan, No. 142 Zhongshan Road of Kunshan City, Jiangsu, China
| | - Jianbo Fan
- Department of Orthopedics, Affiliated Hospital 2 of Nantong University, Jiangsu, China
| |
Collapse
|
10
|
Dong W, Dong Q, Ding H. Ophiopogonin B induces reactive oxygen species‑dependent apoptosis through the Hippo pathway in nasopharyngeal carcinoma. Mol Med Rep 2021; 24:534. [PMID: 34080657 PMCID: PMC8170196 DOI: 10.3892/mmr.2021.12173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignant tumor in South China and is characterized by a high death rate. Ophiopogonin B (OP‑B) is a bioactive component of Radix Ophiopogon japonicus, which is frequently used in traditional Chinese medicine to treat cancer. The present study aimed to examine the anti‑cancer properties of OP‑B on NPC cells. Cell viability and cell proliferation were measured using MTT and EdU assays. Flow cytometry was used to measure cell apoptosis, reactive oxygen species and mitochondrial membrane potential. Western blotting was used to investigate the expression of apoptosis and Hippo signaling pathway proteins. OP‑B inhibited the proliferation of NPC cells by inducing apoptosis and disturbing the mitochondrial integrity. OP‑B enhanced ROS accumulation. In addition, OP‑B promoted the expression of mammalian STE20‑like kinase 1, large tumor suppressor 1 and phosphorylated yes‑associated protein (YAP) and suppressed the expression of YAP and transcriptional enhanced associate domain in NPC cells. OP‑B increased the expression of forkhead box transcription factor O1 in the nuclear fraction. In conclusion, OP‑B has therapeutic potential and feasibility in the development of novel YAP inhibitors for NPC.
Collapse
Affiliation(s)
- Wenhui Dong
- Department of Otorhinolaryngology, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Qing Dong
- Department of Operating Room, Weifang Yidu Central Hospital, Weifang, Shandong 262500, P.R. China
| | - Hairui Ding
- Department of Emergency, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
11
|
Therapeutic potential of AMPK signaling targeting in lung cancer: Advances, challenges and future prospects. Life Sci 2021; 278:119649. [PMID: 34043989 DOI: 10.1016/j.lfs.2021.119649] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Lung cancer (LC) is a leading cause of death worldwide with high mortality and morbidity. A wide variety of risk factors are considered for LC development such as smoking, air pollution and family history. It appears that genetic and epigenetic factors are also potential players in LC development and progression. AMP-activated protein kinase (AMPK) is a signaling pathway with vital function in inducing energy balance and homeostasis. An increase in AMP:ATP and ADP:ATP ratio leads to activation of AMPK signaling by upstream mediators such as LKB1 and CamKK. Dysregulation of AMPK signaling is a common finding in different cancers, particularly LC. AMPK activation can significantly enhance LC metastasis via EMT induction. Upstream mediators such as PLAG1, IMPAD1, and TUFM can regulate AMPK-mediated metastasis. AMPK activation can promote proliferation and survival of LC cells via glycolysis induction. In suppressing LC progression, anti-tumor compounds including metformin, ginsenosides, casticin and duloxetine dually induce/inhibit AMPK signaling. This is due to double-edged sword role of AMPK signaling in LC cells. Furthermore, AMPK signaling can regulate response of LC cells to chemotherapy and radiotherapy that are discussed in the current review.
Collapse
|
12
|
Zhou S, Wen H, Han X, Li H. Phillygenin protects against osteoarthritis by repressing inflammation via PI3K/Akt/NF-κB signaling: In vitro and vivo studies. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
13
|
He J, Wei W, Yang Q, Wang Y. Retracted: Phillygenin Exerts In Vitro and In Vivo Antitumor Effects in Drug-Resistant Human Esophageal Cancer Cells by Inducing Mitochondrial-Mediated Apoptosis, ROS Generation, and Inhibition of the Nuclear Factor kappa B NF-κB Signalling Pathway. Med Sci Monit 2021; 27:e932333. [PMID: 33762558 PMCID: PMC8009254 DOI: 10.12659/msm.932333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jiantao He
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China (mainland)
| | - Wei Wei
- Department of Emergency, Shanghai Tenth People's Hospital, Shanghai, China (mainland)
| | - Qingbo Yang
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China (mainland)
| | - Yiling Wang
- Department of Thoracic Surgery, Shanghai Tenth People's Hospital, Shanghai, China (mainland)
| |
Collapse
|
14
|
Akimov OY, Kostenko VO. Role of NF-κB transcriptional factor activation during chronic fluoride intoxication in development of oxidative-nitrosative stress in rat's gastric mucosa. J Trace Elem Med Biol 2020; 61:126535. [PMID: 32344277 DOI: 10.1016/j.jtemb.2020.126535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 11/19/2022]
Abstract
Fluoride compounds are known as hazardous environmental pollutants that can enter the body with drinking water. Chronic exposure to fluoride leads to development of oxidative stress and can lead to activation of nuclear factor κB (NF-κB). The aim of this work is to clarify the role of NF-kB activation in production of reactive nitrogen and oxygen species, activity of antioxidant enzymes and intensity of lipid peroxidation (LPO) in gastric mucosa of rats during chronic fluoride intoxication. MATERIALS AND METHODS We carried out the study on 18 mature male rats of the Wistar line. The animals were divided into 3 groups: control animals (6), group of chronic fluoride intoxication (6), and animals (6), which received the NF-κB inhibitor, namely ammonium pyrrolidine dithiocarbamate (PDTC) in a dose of 76 mg / kg (iNF-κB group) during modeling of chronic fluoride intoxication. To assess the development of oxidative stress we studied superoxide production (O2-), activity of superoxide dismutase (SOD), catalase (CAT) and concentration of free malondialdehyde (MDA). We also assessed NO production and concentration of its metabolites (peroxynitrite, nitrosilated thiol groups, nitrites). RESULTS Chronic fluoride intoxication leads to NO hyperproduction with subsequent increase in concentration of its later metabolites (peroxynitrite, nitrosilated thiol groups, nitrites). Production of O2- increases, SOD activity decreases, CAT activity increases and MDA concentration also increases. Inhibition of NF-kB activation by PDTC normalizes the parameters studied. CONCLUSIONS Activation of NF-κB during chronic fluoride intoxication leads to the development of hyperproduction of NO and development of oxidative-nitrosative stress.
Collapse
Affiliation(s)
- O Ye Akimov
- Ukrainian Medical Stomatological Academy, Department of Pathophysiology, Poltava, Ukraine.
| | - V O Kostenko
- Ukrainian Medical Stomatological Academy, Department of Pathophysiology, Poltava, Ukraine
| |
Collapse
|
15
|
Li H, Chen M, Yang Z, Wang Q, Wang J, Jin D, Yang X, Chen F, Zhou X, Luo K. Phillygenin, a MELK Inhibitor, Inhibits Cell Survival and Epithelial-Mesenchymal Transition in Pancreatic Cancer Cells. Onco Targets Ther 2020; 13:2833-2842. [PMID: 32308417 PMCID: PMC7138621 DOI: 10.2147/ott.s238958] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction Pancreatic cancer (PC) is one of the leading causes of cancer, with the lowest 5-year survival rate of all cancer types. Given the fast metastasis of PC and its resistance to surgery, radiotherapy, chemotherapy, and combinations thereof, it is imperative to develop more effective anti-PC drugs. Phillygenin (PHI) has been reported to exert anti-cancer, anti-bacterial, and anti‐inflammatory properties. However, the mechanism of PHI in the development of PC is still unclear. Methods The cytotoxicity of PHI in pancreatic cancer cells was evaluated by MTT assay, and clonogenic assay was used to test the anti-proliferation of PHI. The pro-apoptotic effect of PHI was detected by flow cytometry analysis. The changes of epithelial–mesenchymal transition (EMT) in pancreatic cancer cells treated with PHI were determined by Western blot. Transwell assay was used to test the migration and invasion of PC cells after treatment with PHI. Molecular docking was used to predict the potential binding site of candidate target with PHI. Results PHI could inhibit the proliferation, migration, and EMT of PC cells (PANC-1 and SW1990) and induce its apoptosis. Analysis of the Cancer Genome Atlas database indicated that elevated MELK levels correlated with poor overall survival (OS) and disease-free survival (DFS) of PC patients. In addition, molecular modeling showed that PHI may potentially target the catalytic domain of maternal embryonic leucine zipper kinase (MELK). Overexpression of MELK muted the anti-PC effects of PHI. Conclusion PHI holds promise as a potent candidate drug for the treatment of PC via targeted MELK.
Collapse
Affiliation(s)
- Hongchun Li
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Miao Chen
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Zhuying Yang
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Qinxian Wang
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Jiesheng Wang
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Dong Jin
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Xiuyun Yang
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Fuxing Chen
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Xiumin Zhou
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| | - Kexue Luo
- Department of Cadre Health, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang, People's Republic of China
| |
Collapse
|
16
|
Hu N, Wang C, Dai X, Zhou M, Gong L, Yu L, Peng C, Li Y. Phillygenin inhibits LPS-induced activation and inflammation of LX2 cells by TLR4/MyD88/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112361. [PMID: 31683033 DOI: 10.1016/j.jep.2019.112361] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/12/2019] [Accepted: 10/26/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional Chinese medicine Forsythiae Fructus is the dried fruit of Forsythia suspensa (Thunb.) Vahl. It is commonly used to clear heat and detoxify, reduce swelling and disperse knot, and evacuate wind and heat. AIM OF THE STUDY Inflammation is involved in liver fibrosis. Phillygenin (PHI) is a kind of lignans extracted and separated from Forsythiae Fructus, which has been reported to have a good anti-inflammatory effect. Therefore, we aimed to explore whether PHI has a therapeutic effect on liver fibrosis caused by inflammation. MATERIALS AND METHODS Firstly, the induction of the LX2 cells inflammatory model and fibrosis model by LPS with different concentrations were studied. Then, high, medium and low doses PHI was given for intervention therapy. The secretion of IL-6, IL-1β and TNF-α inflammatory factors were detected by ELISA kit, and the expression of collagen I and α-SMA was detected by Western blot and RT-qPCR. The possible mechanism of PHI on TLR4/MyD88/NF-κB signal pathway was studied by computer-aided drug design software and the results were further verified by Western blot and RT-qPCR experiments. RESULTS The results showed that LPS could promote the expression of IL-6, IL-1β and TNF-α and the expression of collagen I and α-SMA, indicating that LPS could induce inflammation and fibrosis in LX2 cells. PHI could inhibit LX2 cell activation and fibrotic cytokine expression by inhibiting LPS-induced pro-inflammatory reaction. Molecular docking results showed that PHI could successfully dock with TLR4, MyD88, IKKβ, p65, IκBα, and TAK1 proteins. Subsequently, Western blot and qPCR results further proved that PHI could inhibit the proteins expression in TLR4/MyD88/NF-κB signal pathway which were consistent with the molecular docking results. CONCLUSION PHI can inhibit LPS-induced pro-inflammatory reaction and LX2 cell activation through TLR4/MyD88/NF-κB signaling pathway, thereby inhibiting liver fibrosis.
Collapse
Affiliation(s)
- Naihua Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Cheng Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Xuyang Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Mengting Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Lihong Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Lingyuan Yu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China.
| | - Yunxia Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China.
| |
Collapse
|
17
|
Guo W, Tan HY, Chen F, Wang N, Feng Y. Targeting Cancer Metabolism to Resensitize Chemotherapy: Potential Development of Cancer Chemosensitizers from Traditional Chinese Medicines. Cancers (Basel) 2020; 12:404. [PMID: 32050640 PMCID: PMC7072159 DOI: 10.3390/cancers12020404] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/22/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer is a common and complex disease with high incidence and mortality rates, which causes a severe public health problem worldwide. As one of the standard therapeutic approaches for cancer therapy, the prognosis and outcome of chemotherapy are still far from satisfactory due to the severe side effects and increasingly acquired resistance. The development of novel and effective treatment strategies to overcome chemoresistance is urgent for cancer therapy. Metabolic reprogramming is one of the hallmarks of cancer. Cancer cells could rewire metabolic pathways to facilitate tumorigenesis, tumor progression, and metastasis, as well as chemoresistance. The metabolic reprogramming may serve as a promising therapeutic strategy and rekindle the research enthusiasm for overcoming chemoresistance. This review focuses on emerging mechanisms underlying rewired metabolic pathways for cancer chemoresistance in terms of glucose and energy, lipid, amino acid, and nucleotide metabolisms, as well as other related metabolisms. In particular, we highlight the potential of traditional Chinese medicine as a chemosensitizer for cancer chemotherapy from the metabolic perspective. The perspectives of metabolic targeting to chemoresistance are also discussed. In conclusion, the elucidation of the underlying metabolic reprogramming mechanisms by which cancer cells develop chemoresistance and traditional Chinese medicines resensitize chemotherapy would provide us a new insight into developing promising therapeutics and scientific evidence for clinical use of traditional Chinese medicine as a chemosensitizer for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 00000, China
| |
Collapse
|
18
|
Sun LR, Zhou W, Zhang HM, Guo QS, Yang W, Li BJ, Sun ZH, Gao SH, Cui RJ. Modulation of Multiple Signaling Pathways of the Plant-Derived Natural Products in Cancer. Front Oncol 2019; 9:1153. [PMID: 31781485 PMCID: PMC6856297 DOI: 10.3389/fonc.2019.01153] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/16/2019] [Indexed: 12/24/2022] Open
Abstract
Natural compounds are highly effective anticancer chemotherapeutic agents, and the targets of plant-derived anticancer agents have been widely reported. In this review, we focus on the main signaling pathways of apoptosis, proliferation, invasion, and metastasis that are regulated by polyphenols, alkaloids, saponins, and polysaccharides. Alkaloids primarily affect apoptosis-related pathways, while polysaccharides primarily target pathways related to proliferation, invasion, and metastasis. Other compounds, such as flavonoids and saponins, affect all of these aspects. The association between compound structures and signaling pathways may play a critical role in drug discovery.
Collapse
Affiliation(s)
- Li-Rui Sun
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Wei Zhou
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Hong-Mei Zhang
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Qiu-Shi Guo
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bing-Jin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Zhi-Hui Sun
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Shuo-Hui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ran-Ji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|