1
|
Zhang Z, Li M, Zhang X, Zhou F. Novel Strategies for Tumor Treatment: Harnessing ROS-Inducing Active Ingredients from Traditional Chinese Medicine Through Multifunctional Nanoformulations. Int J Nanomedicine 2024; 19:9659-9688. [PMID: 39309188 PMCID: PMC11416109 DOI: 10.2147/ijn.s479212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Reactive oxygen species (ROS) encompass a diverse group of chemically reactive molecules or ions distinguished by their substantial oxidative potential. Empirical studies have shown that the targeted administration of high toxic concentrations of ROS can effectively induce tumor cell death in various types. Numerous bioactive ingredients derived from traditional Chinese medicine (TCM), recognized for their ROS-inducing properties, have demonstrated significant anti-tumor activity. Nonetheless, their clinical application has been hindered by challenges such as low solubility, limited bioavailability, and poor selectivity. Multifunctional nanoformulations possess the potential to overcome these challenges and enhance the anticancer efficacy of ROS-inducing active compounds. Through extensive searches of various academic databases and a thorough review and screening of relevant literature, this study aims to systematically summarize and generalize multiple active ingredients in TCM that induce ROS generation, along with their multifunctional nanoformulations, from various perspectives. The objective is to provide new insights and references for fundamental cancer research and clinical treatments. Furthermore, we acknowledge that although numerous active ingredients and their nanoformulations in TCM have demonstrated ROS-inducing and anti-tumor potentials, potentially offering novel strategies for tumor therapy, the underlying mechanisms require further comprehensive investigation.
Collapse
Affiliation(s)
- Zhengguang Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Xiaolong Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Fuqiong Zhou
- Central Laboratory, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
2
|
Sun Y, Li Q, Huang Y, Yang Z, Li G, Sun X, Gu X, Qiao Y, Wu Q, Xie T, Sui X. Natural products for enhancing the sensitivity or decreasing the adverse effects of anticancer drugs through regulating the redox balance. Chin Med 2024; 19:110. [PMID: 39164783 PMCID: PMC11334420 DOI: 10.1186/s13020-024-00982-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024] Open
Abstract
Redox imbalance is reported to play a pivotal role in tumorigenesis, cancer development, and drug resistance. Severe oxidative damage is a general consequence of cancer cell responses to treatment and may cause cancer cell death or severe adverse effects. To maintain their longevity, cancer cells can rescue redox balance and enter a state of resistance to anticancer drugs. Therefore, targeting redox signalling pathways has emerged as an attractive and prospective strategy for enhancing the efficacy of anticancer drugs and decreasing their adverse effects. Over the past few decades, natural products (NPs) have become an invaluable source for developing new anticancer drugs due to their high efficacy and low toxicity. Increasing evidence has demonstrated that many NPs exhibit remarkable antitumour effects, whether used alone or as adjuvants, and are emerging as effective approaches to enhance sensitivity and decrease the adverse effects of conventional cancer therapies by regulating redox balance. Among them are several novel anticancer drugs based on NPs that have entered clinical trials. In this review, we summarize the synergistic anticancer effects and related redox mechanisms of the combination of NPs with conventional anticancer drugs. We believe that NPs targeting redox regulation will represent promising novel candidates and provide prospects for cancer treatment in the future.
Collapse
Affiliation(s)
- Yitian Sun
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Qinyi Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yufei Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Zijing Yang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Guohua Li
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiaoyu Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiaoqing Gu
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yunhao Qiao
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| | - Tian Xie
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Xinbing Sui
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
3
|
Situmorang PC, Ilyas S, Nugraha SE, Syahputra RA, Nik Abd Rahman NMA. Prospects of compounds of herbal plants as anticancer agents: a comprehensive review from molecular pathways. Front Pharmacol 2024; 15:1387866. [PMID: 39104398 PMCID: PMC11298448 DOI: 10.3389/fphar.2024.1387866] [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: 02/18/2024] [Accepted: 06/17/2024] [Indexed: 08/07/2024] Open
Abstract
Cancer refers to the proliferation and multiplication of aberrant cells inside the human body, characterized by their capacity to proliferate and infiltrate various anatomical regions. Numerous biochemical pathways and signaling molecules have an impact on the cancer auto biogenesis process. The regulation of crucial cellular processes necessary for cell survival and proliferation, which are triggered by phytochemicals, is significantly influenced by signaling pathways. These pathways or components are regulated by phytochemicals. Medicinal plants are a significant reservoir of diverse anticancer medications employed in chemotherapy. The anticancer effects of phytochemicals are mediated by several methods, including induction of apoptosis, cessation of the cell cycle, inhibition of kinases, and prevention of carcinogenic substances. This paper analyzes the phytochemistry of seven prominent plant constituents, namely, alkaloids, tannins, flavonoids, phenols, steroids, terpenoids, and saponins, focusing on the involvement of the MAPK/ERK pathway, TNF signaling, death receptors, p53, p38, and actin dynamics. Hence, this review has examined a range of phytochemicals, encompassing their structural characteristics and potential anticancer mechanisms. It has underscored the significance of plant-derived bioactive compounds in the prevention of cancer, utilizing diverse molecular pathways. In addition, this endeavor also seeks to incentivize scientists to carry out clinical trials on anticancer medications derived from plants.
Collapse
Affiliation(s)
- Putri Cahaya Situmorang
- Study Program of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Syafruddin Ilyas
- Study Program of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Sony Eka Nugraha
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Nik Mohd Afizan Nik Abd Rahman
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
4
|
Huang W, Zhang J, Miao C, Ying H, Zhang X, Song M, Cui Y, Wang X, Li Y, Cheng P. Aflatoxin B1-Induced Testosterone Biosynthesis Disorder via the ROS/AMPK Signaling Pathway in Male Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5955-5965. [PMID: 38451160 DOI: 10.1021/acs.jafc.3c08769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The worldwide prevalence of Aflatoxin B1 (AFB1), which contaminates feedstock and food, is on the rise. AFB1 inhibits testosterone (T) biosynthesis, but the mechanism is not yet clear. By establishing in vivo and in vitro models, this study found the number of Leydig cells (LCs), T content, and the expression of T biosynthesis key enzymes were suppressed after AFB1 treatment. AFB1 exposure also increased reactive oxygen species (ROS) and promoted mitochondrial injury and mitochondrial pathway apoptosis. Moreover, the AMPK signaling pathway was activated, and using an AMPK inhibitor relieved apoptosis and the suppressed T biosynthesis key enzymes of LCs caused by AFB1 through regulating downstream p53 and Nur77. Additionally, adding ROS intervention could inhibit AMPK activation and alleviate the decreased T content caused by AFB1. In summary, AFB1 promotes the apoptosis of LCs and inhibits T biosynthesis key enzyme expression via activating the ROS/AMPK signaling pathway, which eventually leads to T synthesis disorder.
Collapse
Affiliation(s)
- Wanyue Huang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Jialu Zhang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Chenjiao Miao
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Huihui Ying
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Xuliang Zhang
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Miao Song
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yilong Cui
- College of Animal Science and Technology, Inner Mongolia Minzu University, 028000 Tongliao, China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Yanfei Li
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ping Cheng
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| |
Collapse
|
5
|
Feng Y, An Q, Zhao Z, Wu M, Yang C, Liang W, Xu X, Jiang T, Zhang G. Beta-elemene: A phytochemical with promise as a drug candidate for tumor therapy and adjuvant tumor therapy. Biomed Pharmacother 2024; 172:116266. [PMID: 38350368 DOI: 10.1016/j.biopha.2024.116266] [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: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND β-Elemene (IUPAC name: (1 S,2 S,4 R)-1-ethenyl-1-methyl-2,4-bis(prop-1-en-2-yl) cyclohexane), is a natural compound found in turmeric root. Studies have demonstrated its diverse biological functions, including its anti-tumor properties, which have been extensively investigated. However, these have not yet been reviewed. The aim of this review was to provide a comprehensive summary of β-elemene research, with respect to disease treatment. METHODS β-Elemene-related articles were found in PubMed, ScienceDirect, and Google Scholar databases to systematically summarize its structure, pharmacokinetics, metabolism, and pharmacological activity. We also searched the Traditional Chinese Medicine System Pharmacology database for therapeutic targets of β-elemene. We further combined these targets with the relevant literature for KEGG and GO analyses. RESULTS Studies on the molecular mechanisms underlying β-elemene activity indicate that it regulates multiple pathways, including STAT3, MAPKs, Cyclin-dependent kinase 1/cyclin B, Notch, PI3K/AKT, reactive oxygen species, METTL3, PTEN, p53, FAK, MMP, TGF-β/Smad signaling. Through these molecular pathways, β-elemene has been implicated in tumor cell proliferation, apoptosis, migration, and invasion and improving the immune microenvironment. Additionally, β-elemene increases chemotherapeutic drug sensitivity and reverses resistance by inhibiting DNA damage repair and regulating pathways including CTR1, pak1, ERK1/2, ABC transporter protein, Prx-1 and ERCC-1. Nonetheless, owing to its lipophilicity and low bioavailability, additional structural modifications could improve the efficacy of this drug. CONCLUSION β-Elemene exhibits low toxicity with good safety, inhibiting various tumor types via diverse mechanisms in vivo and in vitro. When combined with chemotherapeutic drugs, it enhances efficacy, reduces toxicity, and improves tumor killing. Thus, β-elemene has vast potential for research and development.
Collapse
Affiliation(s)
- Yewen Feng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Qingwen An
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Zhengqi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Mengting Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Chuqi Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - WeiYu Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Xuefei Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| |
Collapse
|
6
|
Cao LY, Xu JY, Zhuo XT, Zhang W, Wei LJ, Dong JH, Bai RR, Wang X, Jiang YY, Wang YJ, Ye XY, Xie T, Huang ZH. 2,2'-((1R,3R,4S)-4-methyl-4-vinylcyclohexane-1,3-diyl) bis(prop-2-en-1-amine), a bisamino derivative of β-Elemene, inhibits glioblastoma growth through downregulation of YAP signaling. Am J Cancer Res 2022; 12:5484-5499. [PMID: 36628286 PMCID: PMC9827083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/20/2022] [Indexed: 01/12/2023] Open
Abstract
β-Elemene, a compound extracted from Chinese herb Curcuma wenyujin, has been demonstrated with antitumor effects in various cancers, including glioblastoma (GBM), a primary brain tumor with high morbidity and mortality. In this study, we reported a bisamino derivative of β-Elemene, 2, 2'-((1R, 3R, 4S)-4-methyl-4-vinylcyclohexane-1, 3-diyl) bis(prop-2-en-1-amine) (compound 1), displayed a better anti-GBM effect than β-Elemene with lower concentration. GBM cell lines (C6 and U87) were treated with compound 1 and subsequently analyzed by several assays. Compound 1 significantly inhibited the migration of C6 and U87 cells based on wound healing assay, transwell assay and inverted migration assay. Furthermore, colony formation assay, immunostaining and flow cytometry assays revealed that compound 1 significantly inhibited the proliferation of GBM cells. In addition, compound 1 induced the apoptosis of GBM cells. Mechanistically, we found Yes-associated protein (YAP) was down-regulated in compound 1-treated GBM cells, and the overexpression of YAP partially rescued the anti-GBM effects of compound 1. Finally, compound 1 suppresses the GBM growth in xenograft model through inactivation YAP signaling. Taken together, these results reveal that a novel derivative of β-Elemene, compound 1, exhibits more potent anti-GBM activity than β-Elemene through inactivating YAP signaling pathway, which will provide novel strategies for the treatment of GBM.
Collapse
Affiliation(s)
- Li-Ying Cao
- Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Jia-Yun Xu
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Xiao-Tao Zhuo
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Wei Zhang
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Li-Jia Wei
- Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Jian-Hong Dong
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Ren-Ren Bai
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Xin Wang
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Yuan-Yuan Jiang
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Yong-Jie Wang
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Xiang-Yang Ye
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| | - Zhi-Hui Huang
- School of Pharmacy, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou Normal UniversityHangzhou 311121, Zhejiang, China
| |
Collapse
|
7
|
Li B, Shao H, Gao L, Li H, Sheng H, Zhu L. Nano-drug co-delivery system of natural active ingredients and chemotherapy drugs for cancer treatment: a review. Drug Deliv 2022; 29:2130-2161. [PMID: 35815678 PMCID: PMC9275501 DOI: 10.1080/10717544.2022.2094498] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy drugs have been used for a long time in the treatment of cancer, but serious side effects are caused by the inability of the drug to be solely delivered to the tumor when treating cancer with chemotherapy. Natural products have attracted more and more attention due to the antitumor effect in multiple ways, abundant resources and less side effects. Therefore, the combination of natural active ingredients and chemotherapy drugs may be an effective antitumor strategy, which can inhibit the growth of tumor and multidrug resistance, reduce side effects of chemotherapy drugs. Nano-drug co-delivery system (NDCDS) can play an important role in the combination of natural active ingredients and chemotherapy drugs. This review provides a comprehensive summary of the research status and application prospect of nano-delivery strategies for the combination of natural active ingredients and chemotherapy drugs, aiming to provide a basis for the development of anti-tumor drugs.
Collapse
Affiliation(s)
- Bingqian Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huili Shao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huan Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
8
|
Gao Q, Feng J, Liu W, Wen C, Wu Y, Liao Q, Zou L, Sui X, Xie T, Zhang J, Hu Y. Opportunities and challenges for co-delivery nanomedicines based on combination of phytochemicals with chemotherapeutic drugs in cancer treatment. Adv Drug Deliv Rev 2022; 188:114445. [PMID: 35820601 DOI: 10.1016/j.addr.2022.114445] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 02/08/2023]
Abstract
The therapeutic limitations such as insufficient efficacy, drug resistance, metastasis, and undesirable side effects are frequently caused by the long duration monotherapy based on chemotherapeutic drugs. multiple combinational anticancer strategies such as nucleic acids combined with chemotherapeutic agents, chemotherapeutic combinations, chemotherapy and tumor immunotherapy combinations have been embraced, holding great promise to counter these limitations, while still taking including some potential risks. Nowadays, an increasing number of research has manifested the anticancer effects of phytochemicals mediated by modulating cancer cellular events directly as well as the tumor microenvironment. Specifically, these natural compounds exhibited suppression of cancer cell proliferation, apoptosis, migration and invasion of cancer cells, P-glycoprotein inhibition, decreasing vascularization and activation of tumor immunosuppression. Due to the low toxicity and multiple modulation pathways of these phytochemicals, the combination of chemotherapeutic agents with natural compounds acts as a novel approach to cancer therapy to increase the efficiency of cancer treatments as well as reduce the adverse consequences. In order to achieve the maximized combination advantages of small-molecule chemotherapeutic drugs and natural compounds, a variety of functional nano-scaled drug delivery systems, such as liposomes, host-guest supramolecules, supramolecules, dendrimers, micelles and inorganic systems have been developed for dual/multiple drug co-delivery. These co-delivery nanomedicines can improve pharmacokinetic behavior, tumor accumulation capacity, and achieve tumor site-targeting delivery. In that way, the improved antitumor effects through multiple-target therapy and reduced side effects by decreasing dose can be implemented. Here, we present the synergistic anticancer outcomes and the related mechanisms of the combination of phytochemicals with small-molecule anticancer drugs. We also focus on illustrating the design concept, and action mechanisms of nanosystems with co-delivery of drugs to synergistically improve anticancer efficacy. In addition, the challenges and prospects of how these insights can be translated into clinical benefits are discussed.
Collapse
Affiliation(s)
- Quan Gao
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiao Feng
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wencheng Liu
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chengyong Wen
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Xinbing Sui
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Tian Xie
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China.
| |
Collapse
|
9
|
Clinical Value Analysis of Xiaozheng Decoction Combined with Bladder Perfusion for Postoperative Treatment of Bladder Cancer and Its Effect on Serum miR-143 and miR-92a. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:8177674. [PMID: 35368948 PMCID: PMC8970859 DOI: 10.1155/2022/8177674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 01/16/2023]
Abstract
Objective To study the clinical value of Xiaozheng decoction combined with bladder perfusion treating bladder cancer after the operation and its effect on serum miR-143 and miR-92a. Methods The patients in the control group were treated with gemcitabine bladder instillation, and patients in the study group were treated with the combination of gemcitabine bladder instillation + Xiaozheng decoction. The clinical efficacy, postoperative adverse effects, and recurrence between the two groups were compared. miR-143 and miR-92a levels, immune function levels, and tumor factor levels were compared before and after treatment. The relationship between patient prognosis and miR-143/miR-92a expression was analyzed. Results The overall effective rate of treatment in the study group (86.67%) was significantly increased, and the occurrence of adverse reactions and recurrence were significantly decreased (P < 0.05). After treatment, serum miR-143 and miR-92a levels, CD4+, CD4+/CD8+, and NK levels increased in both groups (P<0.05). CD8+ levels, BTA, NMP, and UBC levels decreased in both groups (P < 0.05). Analysis of survival results indicated that the two-year survival rates of patients with miR-143 and miR-92a high expressions were significantly higher than patients with low expressions (P < 0.05). Conclusion The efficacy of Xiaozheng decoction combined with bladder perfusion in treating postoperative patients with bladder cancer was significant, which could reduce the incidence of adverse reactions and postoperative recurrence rate, improve serum tumor marker levels, and enhance patients' immunity with a good prognosis.
Collapse
|
10
|
Zhang Y, Cheng J, Li J, He J, Li X, Xu F. The GLP-1R Agonist Exendin-4 Attenuates Hyperglycemia-Induced Chemoresistance in Human Endometrial Cancer Cells Through ROS-Mediated Mitochondrial Pathway. Front Oncol 2022; 11:793530. [PMID: 34988025 PMCID: PMC8721044 DOI: 10.3389/fonc.2021.793530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/01/2021] [Indexed: 12/23/2022] Open
Abstract
This study aimed to assess the effects of the antidiabetic drug Exendin-4 (Exe-4), a GLP-1 receptor agonist, on the response of human endometrial cancer cells to chemotherapy under high glucose (HG) conditions. Cell viability was detected using a cell counting kit (CCK)-8. Cell apoptosis and reactive oxygen species (ROS) levels were measured by flow cytometry. Gene expression was evaluated by real-time PCR and immunoblotting. The chemotherapeutic drug cisplatin (DDP) dose-dependently inhibited both human endometrial adenocarcinoma Ishikawa and HEC1B cells, a response reversed by HG. Meanwhile, Exe-4 attenuated hyperglycemia’s effect by elevating intracellular lactate dehydrogenase (LDH) and ROS production. Similarly, DDP-induced elevation of intracellular rhodamine123 was attenuated by HG, and Exe-4 reversed HG’s impact. The chemoresistance genes multidrug resistance-associated protein 1 (MRP1) and P-glycoprotein (Pgp) were upregulated. At the same time, topoisomerase II (TOPO II) was downregulated under HG conditions, suggesting HG-induced chemoresistance. Exe-4 did not significantly influence the above genes. DDP downregulated Bcl-2 and Bcl-XL and upregulated Bax, cytosolic cytochrome c, and PARP under normal glucose (NG) versus HG conditions, and Exe-4 attenuated these effects. Upstream of Bax/Bcl, acetylated P53 was upregulated by DDP and downregulated by HG, whose effect was reversed by Exe-4. DPP treatment significantly induced apoptosis and cell cycle arrest in the S phase under NG, and HG reduced these effects. Prolonged exposure to HG induces DDP chemoresistance in human endometrial cancer cells but is alleviated by Exe-4.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Juan Cheng
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Jing Li
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Junxian He
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Xiaomao Li
- Department of Gynecology, Third Affiliated Hospital, Sun-Yet Sen University, Guangzhou, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| |
Collapse
|
11
|
Bai Y, Sha J, Okui T, Moriyama I, Ngo HX, Tatsumi H, Kanno T. The Epithelial-Mesenchymal Transition Influences the Resistance of Oral Squamous Cell Carcinoma to Monoclonal Antibodies via Its Effect on Energy Homeostasis and the Tumor Microenvironment. Cancers (Basel) 2021; 13:5905. [PMID: 34885013 PMCID: PMC8657021 DOI: 10.3390/cancers13235905] [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: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a major type of cancer that accounts for over 90% of all oral cancer cases. Recently developed evidence-based therapeutic regimens for OSCC based on monoclonal antibodies (mAbs), such as cetuximab, pembrolizumab, and nivolumab, have attracted considerable attention worldwide due to their high specificity, low toxicity, and low rates of intolerance. However, the efficacy of those three mAbs remains poor because of the low rate of responders and acquired resistance within a short period of time. The epithelial-mesenchymal transition (EMT) process is fundamental for OSCC growth and metastasis and is also responsible for the poor response to mAbs. During EMT, cancer cells consume abundant energy substrates and create an immunosuppressive tumor microenvironment to support their growth and evade T cells. In this review, we provide an overview of the complex roles of major substrates and signaling pathways involved in the development of therapeutic resistance in OSCC. In addition, we summarize potential therapeutic strategies that may help overcome this resistance. This review aims to help oral oncologists and researchers aiming to manage OSCC and establish new treatment modalities.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Jingjing Sha
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Ichiro Moriyama
- Department of Medical Oncology/Innovative Cancer Center, Shimane University Hospital, Izumo, Shimane 693-8501, Japan;
| | - Huy Xuan Ngo
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Hiroto Tatsumi
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Takahiro Kanno
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| |
Collapse
|
12
|
Tan T, Li J, Luo R, Wang R, Yin L, Liu M, Zeng Y, Zeng Z, Xie T. Recent Advances in Understanding the Mechanisms of Elemene in Reversing Drug Resistance in Tumor Cells: A Review. Molecules 2021; 26:5792. [PMID: 34641334 PMCID: PMC8510449 DOI: 10.3390/molecules26195792] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
Malignant tumors are life-threatening, and chemotherapy is one of the common treatment methods. However, there are often many factors that contribute to the failure of chemotherapy. The multidrug resistance of cancer cells during chemotherapy has been reported, since tumor cells' sensitivity decreases over time. To overcome these problems, extensive studies have been conducted to reverse drug resistance in tumor cells. Elemene, an extract of the natural drug Curcuma wenyujin, has been found to reverse drug resistance and sensitize cancer cells to chemotherapy. Mechanisms by which elemene reverses tumor resistance include inhibiting the efflux of ATP binding cassette subfamily B member 1(ABCB1) transporter, reducing the transmission of exosomes, inducing apoptosis and autophagy, regulating the expression of key genes and proteins in various signaling pathways, blocking the cell cycle, inhibiting stemness, epithelial-mesenchymal transition, and so on. In this paper, the mechanisms of elemene's reversal of drug resistance are comprehensively reviewed.
Collapse
Affiliation(s)
- Tiantian Tan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Jie Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruhua Luo
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Rongrong Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Liyan Yin
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mengmeng Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Yiying Zeng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhaowu Zeng
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; (T.T.); (J.L.); (R.L.); (R.W.); (L.Y.); (M.L.)
- Key Laboratory of Element Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
13
|
Protective Effect of Penetratin Analogue-Tagged SOD1 on Cisplatin-Induced Nephrotoxicity through Inhibiting Oxidative Stress and JNK/p38 MAPK Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5526053. [PMID: 34471466 PMCID: PMC8405295 DOI: 10.1155/2021/5526053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/02/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022]
Abstract
Copper/zinc superoxide dismutase (SOD1) can clear cisplatin- (CP-) induced excessive reactive oxygen species (ROS), but exogenous SOD1 cannot enter cells because of its low biomembrane permeability. Cell-penetrating peptides (CPPs) can rapidly cross plasma membranes. This study is aimed at identifying an efficient and stable CPP-SOD1 and investigating its effects on CP-induced nephrotoxicity. We recombined SOD1 with 14 different CPPs and purified them using an NTA-Ni2+ column. In in vitro experiments, CPPs-SOD1 cell membrane penetration ability and JNK/p38 MAPK signaling pathway were evaluated using Western blotting. ROS production, mitochondrial membrane potential (MMP), and cell apoptosis were determined using flow cytometry and immunofluorescence staining in VERO and HK-2 cells. For in vivo experiments, mice were administered PSF-SOD1 for 2 h before cotreatment with a single CP injection for an additional 4 days. Blood and kidney samples were collected for renal function assessment (creatinine, urea nitrogen, histopathology, TUNEL assay, and JNK/p38 MAPK signaling pathway). Compared with TAT-SOD1, we found that PSF-SOD1 is more efficient at crossing the cell membrane and is stable after transduction into cells. Pretreatment with PSF-SOD1 inhibited CP-induced apoptosis, ROS generation, and JNK/p38 MAPK activation and restored CP-induced MMP loss in VERO and HK-2 kidney cells. Treatment of mice with PSF-SOD1 inhibited CP-induced serum creatinine, blood urea nitrogen elevation, and JNK/p38 MAPK activation. H&E staining and TUNEL assay indicated that kidney tissue damage was alleviated following PSF-SOD1 pretreatment. Overall, PSF-SOD1 ameliorated CP-induced renal damage by partially reducing oxidative stress and cell apoptosis by regulating JNK/p38 MAPK signaling pathway and might be a better cytoprotective agent than TAT-SOD1.
Collapse
|
14
|
Mirzaei S, Hushmandi K, Zabolian A, Saleki H, Torabi SMR, Ranjbar A, SeyedSaleh S, Sharifzadeh SO, Khan H, Ashrafizadeh M, Zarrabi A, Ahn KS. Elucidating Role of Reactive Oxygen Species (ROS) in Cisplatin Chemotherapy: A Focus on Molecular Pathways and Possible Therapeutic Strategies. Molecules 2021; 26:2382. [PMID: 33921908 PMCID: PMC8073650 DOI: 10.3390/molecules26082382] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
The failure of chemotherapy is a major challenge nowadays, and in order to ensure effective treatment of cancer patients, it is of great importance to reveal the molecular pathways and mechanisms involved in chemoresistance. Cisplatin (CP) is a platinum-containing drug with anti-tumor activity against different cancers in both pre-clinical and clinical studies. However, drug resistance has restricted its potential in the treatment of cancer patients. CP can promote levels of free radicals, particularly reactive oxygen species (ROS) to induce cell death. Due to the double-edged sword role of ROS in cancer as a pro-survival or pro-death mechanism, ROS can result in CP resistance. In the present review, association of ROS with CP sensitivity/resistance is discussed, and in particular, how molecular pathways, both upstream and downstream targets, can affect the response of cancer cells to CP chemotherapy. Furthermore, anti-tumor compounds, such as curcumin, emodin, chloroquine that regulate ROS and related molecular pathways in increasing CP sensitivity are described. Nanoparticles can provide co-delivery of CP with anti-tumor agents and by mediating photodynamic therapy, and induce ROS overgeneration to trigger CP sensitivity. Genetic tools, such as small interfering RNA (siRNA) can down-regulate molecular pathways such as HIF-1α and Nrf2 to promote ROS levels, leading to CP sensitivity. Considering the relationship between ROS and CP chemotherapy, and translating these findings to clinic can pave the way for effective treatment of cancer patients.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran 1477893855, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417466191, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1477893855, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1477893855, Iran
| | - Seyed Mohammad Reza Torabi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1477893855, Iran
| | - Adnan Ranjbar
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1477893855, Iran
| | - SeyedHesam SeyedSaleh
- Student Research Committee, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Seyed Omid Sharifzadeh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1477893855, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul 34956, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Kwang-Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
15
|
Anti-Tumor Drug Discovery Based on Natural Product β-Elemene: Anti-Tumor Mechanisms and Structural Modification. Molecules 2021; 26:molecules26061499. [PMID: 33801899 PMCID: PMC7998186 DOI: 10.3390/molecules26061499] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022] Open
Abstract
Natural products are important sources for drug discovery, especially anti-tumor drugs. β-Elemene, the prominent active ingredient extract from the rhizome of Curcuma wenyujin, is a representative natural product with broad anti-tumor activities. The main molecular mechanism of β-elemene is to inhibit tumor growth and proliferation, induce apoptosis, inhibit tumor cell invasion and metastasis, enhance the sensitivity of chemoradiotherapy, regulate the immune system, and reverse multidrug resistance (MDR). Elemene oral emulsion and elemene injection were approved by the China Food and Drug Administration (CFDA) for the treatment of various cancers and bone metastasis in 1994. However, the lipophilicity and low bioavailability limit its application. To discover better β-elemene-derived anti-tumor drugs with satisfying drug-like properties, researchers have modified its structure under the premise of not damaging the basic scaffold structure. In this review, we comprehensively discuss and summarize the potential anti-tumor mechanisms and the progress of structural modifications of β-elemene.
Collapse
|
16
|
Wang H, Zhu J, Jiang L, Shan B, Xiao P, Ai J, Li N, Qi F, Niu S. Mechanism of Heshouwuyin inhibiting the Cyt c/Apaf-1/Caspase-9/Caspase-3 pathway in spermatogenic cell apoptosis. BMC Complement Med Ther 2020; 20:180. [PMID: 32527252 PMCID: PMC7291440 DOI: 10.1186/s12906-020-02904-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/26/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The Chinese herbal compound Heshouwuyin has been shown to downregulate the apoptotic rate of testicular tissue cells in Wistar naturally aging rats, and this effect might be related to the mitochondrial pathway [15]. Apoptotic protease activating factor-1 (Apaf-1) is a major component of the apoptotic complex, which is a key element of the mitochondrial endogenous apoptotic pathway [13]. To further clarify the mechanism of Heshouwuyin in the mitochondrial apoptotic pathway, this study used Apaf-1 as a target to explore the mechanism by which Heshouwuyin inhibits the Apaf-1 pathway of spermatogenic cell apoptosis. METHODS In this study, an aging model of rat spermatogenic cells was established using free radical oxidative damage. Flow cytometry was used to detect the apoptosis rate of germ cells and the inhibitory effect of Heshouwuyin. Apaf-1 was specifically knocked down by siRNA interference technology, and mitochondrial membrane potential was measured. qRT-PCR, Western blotting and immunofluorescence analyses were used to detect the expression of the key genes Cyt c, Caspase-9 and Caspase-3 in the mitochondrial apoptotic pathway of spermatogenic cells. RESULTS Heshouwuyin reduced the mRNA and protein expression levels of Cyt c, Caspase-9 and Caspase-3 in senescent spermatogenic cells. In these cells, the mRNA and protein expression levels of Cyt c did not change significantly after specific knockdown of Apaf-1, and the mRNA and protein expression levels of Caspase-9 and Caspase-3 decreased significantly. This finding indicated that knockdown of Apaf-1 could decrease the mRNA and protein expression levels of the downstream pro-apoptotic genes Caspase-9 and Caspase-3. Although Cyt c was an upstream gene of Apaf-1, knockdown of Apaf-1 had no significant effect on Cyt c expression. CONCLUSION The inhibition of spermatogenic cell apoptosis by Heshouwuyin was closely related to the Cyt c/Apaf-1/Caspase-9/Caspase-3 pathway. The inhibition of apoptosis by Heshouwuyin not only involved the Apaf-1 pathway, but other signaling pathways.
Collapse
Affiliation(s)
- Hongjie Wang
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China.,Affiliated Hospital of Hebei University, Baoding, 071002, Hebei Province, China
| | - Juan Zhu
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China
| | - Liping Jiang
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China
| | - Boying Shan
- Nanbao Development Zone Hospital, Tangshan, 063305, Hebei Province, China
| | - Peihan Xiao
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China
| | - Jiayi Ai
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China
| | - Na Li
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China
| | - Feng Qi
- Baoding No.1 Hospital, Baoding, 071000, Hebei Province, China.
| | - Siyun Niu
- School of Medicine, Hebei University, Baoding, 071002, Hebei Province, China.
| |
Collapse
|
17
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Kalantari M, Mohammadinejad R, Javaheri T, Sethi G. Association of the Epithelial-Mesenchymal Transition (EMT) with Cisplatin Resistance. Int J Mol Sci 2020; 21:E4002. [PMID: 32503307 PMCID: PMC7312011 DOI: 10.3390/ijms21114002] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023] Open
Abstract
Therapy resistance is a characteristic of cancer cells that significantly reduces the effectiveness of drugs. Despite the popularity of cisplatin (CP) as a chemotherapeutic agent, which is widely used in the treatment of various types of cancer, resistance of cancer cells to CP chemotherapy has been extensively observed. Among various reported mechanism(s), the epithelial-mesenchymal transition (EMT) process can significantly contribute to chemoresistance by converting the motionless epithelial cells into mobile mesenchymal cells and altering cell-cell adhesion as well as the cellular extracellular matrix, leading to invasion of tumor cells. By analyzing the impact of the different molecular pathways such as microRNAs, long non-coding RNAs, nuclear factor-κB (NF-ĸB), phosphoinositide 3-kinase-related protein kinase (PI3K)/Akt, mammalian target rapamycin (mTOR), and Wnt, which play an important role in resistance exhibited to CP therapy, we first give an introduction about the EMT mechanism and its role in drug resistance. We then focus specifically on the molecular pathways involved in drug resistance and the pharmacological strategies that can be used to mitigate this resistance. Overall, we highlight the various targeted signaling pathways that could be considered in future studies to pave the way for the inhibition of EMT-mediated resistance displayed by tumor cells in response to CP exposure.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran;
- Kazerun Health Technology Incubator, Shiraz University of Medical Sciences, Shiraz 1433671348, Iran
| | - Mahshad Kalantari
- Department of Genetic Science, Tehran Medical Science Branch, Islamic Azad University, Tehran 19168931813, Iran;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 1355576169, Iran
| | - Tahereh Javaheri
- Health Informatics Lab, Metropolitan College, Boston University, Boston, MA 02215, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
| |
Collapse
|
18
|
Wang Y, Jiang XY, Yu XY. BRD9 controls the oxytocin signaling pathway in gastric cancer via CANA2D4, CALML6, GNAO1, and KCNJ5. Transl Cancer Res 2020; 9:3354-3366. [PMID: 35117701 PMCID: PMC8798819 DOI: 10.21037/tcr.2020.03.67] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/08/2020] [Indexed: 01/26/2023]
Abstract
Background First-line chemotherapeutic agents lead to remarkable activation treatment in cancers, but the side effects of these drugs also damage healthy cells. In some cases, drug resistance to chemotherapeutic agents is induced in cancer cells. The molecular mechanisms underlying such a side effect have been studied in a range of cancer types, yet little is known about how the adverse effects of chemotherapeutic drugs can be diminished by targeting bromodomain-containing protein 9 (BRD9) in gastric cancers. Methods We used two gastric cancer cell lines (MGC-803 and AGS) for comparison. We applied molecular and cellular techniques to measure cell survival and mRNA expression, investigated clinical data in the consensus of The Cancer Genome Atlas, and utilized high-throughput sequencing in MGC-803 cells and AGS cells for global gene expression analysis in inhibiting BRD9 conditions. Results Our studies showed that cancer cells with BRD9 overexpression, MGC-803 cells, were more sensitive to BRD9 inhibitors (i.e., BI9564 or BI7273) than AGS cells. The mechanism of BRD9 was related to the regulation of calcium voltage-gated channel auxiliary subunit alpha2 delta 4 (CANA2D4), calmodulin-like 6 (CALML6), guanine nucleotide binding protein (G protein), alpha activating activity polypeptide O (GNAO1) and Potassium Inwardly Rectifying Channel Subfamily J, Member 5 (KCNJ5) oncogenes in the oxytocin signaling pathway. BRD9 inhibitors could enhance the sensitivity of gastric cancer MGC-803 cells to adriamycin and cisplatin, so we may reduce the dosage of chemotherapeutic agents in curing gastric cancers with BRD9 over expression by combining BI9564 or BI7273 with adriamycin or cisplatin. Conclusions Our study elucidated the feasibility and effectiveness of inhibiting BRD9 to reduce the adverse effects of first-line chemotherapeutic agents in treating gastric cancer with BRD9 overexpression. This study provides a scientific theoretical basis for a chemotherapy regimen in gastric cancer with BRD9 overexpression.
Collapse
Affiliation(s)
- Yuan Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Affiliated Fifth Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xue-Yan Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Affiliated Fifth Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Affiliated Fifth Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|