1
|
Shaw P, Dey Bhowmik A, Gopinatha Pillai MS, Robbins N, Dwivedi SKD, Rao G. Anoikis resistance in Cancer: Mechanisms, therapeutic strategies, potential targets, and models for enhanced understanding. Cancer Lett 2025; 624:217750. [PMID: 40294841 DOI: 10.1016/j.canlet.2025.217750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Anoikis, defined as programmed cell death triggered by the loss of cell-extracellular matrix (ECM) and cell-cell interactions, is crucial for maintaining tissue homeostasis and preventing aberrant cell migration. Cancer cells, however, display anoikis resistance (AR) which in turn enables cancer metastasis. AR results from alterations in apoptotic signaling, metabolic reprogramming, autophagy modulation, and epigenetic changes, allowing cancer cells to survive in detached conditions. In this review we describe the mechanisms underlying both anoikis and AR, focusing on intrinsic and extrinsic pathways, disrupted cell-ECM interactions, and autophagy in cancer. Recent findings (i.e., between 2014 and 2024) on epigenetic regulation of AR and its role in metastasis are discussed. Therapeutic strategies targeting AR, including chemical inhibitors, are highlighted alongside a network analysis of 122 proteins reported to be associated with AR which identifies 53 hub proteins as potential targets. We also evaluate in vitro and in vivo models for studying AR, emphasizing their role in advancing metastasis research. Our overall goal is to guide future studies and therapeutic developments to counter cancer metastasis.
Collapse
Affiliation(s)
- Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Nathan Robbins
- James E. Hurley School of Science and Mathematics, Oklahoma Baptist University, Shawnee, OK, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA.
| |
Collapse
|
2
|
Zhao T, Cai X, Chen H, Wang Z, Bu H, Lin S. Rg3 inhibits hypoxia-induced tumor exosomes from boosting pancreatic cancer vasculogenic mimicry through the HIF-1α/LARS1/mTOR axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156437. [PMID: 39955826 DOI: 10.1016/j.phymed.2025.156437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Pancreatic cancer (PC), characterized by a poor prognosis, can utilize hypoxia to activate vasculogenic mimicry (VM) and facilitate metastasis. Exosomes serve as crucial mediators in this hypoxic environment. Our previous studies demonstrated that Rg3 could counteract VM by modulating the impact of PC-derived exosomes. AIM This study focuses on the carcinogenic mechanism of PC in a hypoxic environment. METHODS Exosomes from PANC-1 and BxPC-3 cells under normoxic or hypoxic conditions were isolated and characterized by western blot (WB) and nanoparticle trafficking analysis. These PC cells' VM potential was assessed through tube formation and WB. Molecular mechanisms were explored using proteomics analysis, bioinformatics analysis, and the gain- and loss-of-function studies, and the efficacy of Rg3 targeting these exosomes was examined in vitro and in vivo. RESULTS Exosomes from PANC-1 and BxPC-3 cells enhanced VM formation in PC cells under hypoxic conditions. Proteomics analysis revealed that these exosomes involved the HIF-1α/LARS1/mTOR axis. Hypoxia-activated HIF-1α led to high expression of LARS1 in PC cell exosomes, which were uptaken by recipient PC cells activating the mTOR signaling and promoting VM formation. Interaction between HIF-1α and LARS1 was further confirmed. In vitro and in vivo experiments demonstrated that Rg3 can diminish VM formation of PC cells triggered by the LARS1/mTOR axis in PC-derived exosomes under hypoxic conditions, improving the therapeutic effect of Rg3. CONCLUSIONS Our findings revealed a novel mechanism through which Rg3 inhibits VM in PC by modulating hypoxia-induced tumor exosomes, offering novel experimental insights for PC treatment.
Collapse
Affiliation(s)
- Tingting Zhao
- School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, PR China
| | - Xufan Cai
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China; Cancer Center, Department of Thoracic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, Zhejiang, PR China
| | - Hui Chen
- Department of hepatobiliary and pancreatic surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Zhaohong Wang
- Department of hepatobiliary and pancreatic surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, PR China
| | - Heqi Bu
- Department of Colorectal Surgery, Beilun District People's Hospital, Ningbo 315800, Zhejiang, PR China
| | - Shengzhang Lin
- School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, PR China.
| |
Collapse
|
3
|
He C, He J. Metabolic reprogramming and signaling adaptations in anoikis resistance: mechanisms and therapeutic targets. Mol Cell Biochem 2025:10.1007/s11010-024-05199-3. [PMID: 39821582 DOI: 10.1007/s11010-024-05199-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025]
Abstract
Anoikis, a form of programmed cell death triggered by detachment from the extracellular matrix (ECM), maintains tissue homeostasis by removing mislocalized or detached cells. Cancer cells, however, have evolved multiple mechanisms to evade anoikis under conditions of ECM detachment, enabling survival and distant metastasis. Studies have identified differentially expressed proteins between suspended and adherent cancer cells, revealing that key metabolic and signaling pathways undergo significant alterations during the acquisition of anoikis resistance. This review explores the regulatory roles of epithelial-mesenchymal transition, cancer stem cell characteristics, metabolic reprogramming, and various signaling pathway alterations in promoting anoikis resistance. And the corresponding reagents and non-coding RNAs that target the aforementioned pathways are reviewed. By discussing the regulatory mechanisms that facilitate anoikis resistance in cancer cells, this review aims to shed light on potential strategies for inhibiting tumor progression and preventing metastasis.
Collapse
Affiliation(s)
- Chao He
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie He
- Department of Nursing, Operating Room, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
4
|
Foglio E, D'Avorio E, Nieri R, Russo MA, Limana F. Epicardial EMT and cardiac repair: an update. Stem Cell Res Ther 2024; 15:219. [PMID: 39026298 PMCID: PMC11264588 DOI: 10.1186/s13287-024-03823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024] Open
Abstract
Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, Latina, Italy
| | - Erica D'Avorio
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy
| | - Riccardo Nieri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Federica Limana
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy.
- Laboratorio di Patologia Cellulare e Molecolare, IRCCS San Raffaele Roma, Rome, Italy.
| |
Collapse
|
5
|
Zou F, Zhang G, Mei G, Zhang H, Xie M, Dan M. CTEN-induced TGF-β1 expression facilitates EMT and enhances paclitaxel resistance in bladder cancer cells. Am J Transl Res 2024; 16:3248-3258. [PMID: 39114729 PMCID: PMC11301497 DOI: 10.62347/qwak3951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/14/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVES To investigate the role of C-terminal tensin-like (CTEN) in mediating chemotherapy resistance via epithelial-mesenchymal transition (EMT) in bladder cancer (BC) cells, through the regulation of transforming growth factor-β1 (TGF-β1) expression. METHODS Lentiviral vectors were used to create CTEN overexpression and knockdown constructs, which were then introduced into paclitaxel-resistant BC cell lines. The effects of CTEN manipulation on cell proliferation and drug sensitivity was assessed using the CCK-8 assay, and apoptosis was evaluated by flow cytometry. The expression levels of CTEN, TGF-β1, and EMT markers were quantified by RT-qPCR and Western blot analysis. The interaction between CTEN and TGF-β1 and its effect on TGF-β1 methylation were studied using bisulfite sequencing PCR and co-immunoprecipitation. RESULTS Overexpression of CTEN in BC cells was associated with decreased paclitaxel efficacy, reduced apoptosis, and elevated levels of TGF-β1 and EMT-related proteins. CTEN was found to bind TGF-β1, inhibiting its methylation and thereby promoting TGF-β1 upregulation. This increase in TGF-β1 expression facilitated the EMT process and enhanced drug resistance in BC cells. CONCLUSIONS The induction of TGF-β1 expression by CTEN promotes EMT and increases chemotherapy resistance in BC cells. Targeting CTEN or the EMT pathway could improve chemosensitivity in treatment-resistant BC, suggesting a novel therapeutic strategy to enhance chemotherapy effectiveness.
Collapse
Affiliation(s)
- Feng Zou
- Department of Urology, The Seventh Affiliated Hospital, Southern Medical UniversityFoshan 528000, Guangdong, China
| | - Guofei Zhang
- Department of Urology, The Seventh Affiliated Hospital, Southern Medical UniversityFoshan 528000, Guangdong, China
| | - Gang Mei
- Department of Orthopedics, The Seventh Affiliated Hospital, Southern Medical UniversityFoshan 528000, Guangdong, China
| | - Huantao Zhang
- Department of Urology Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen UniversityHuizhou 516200, Guangdong, China
| | - Mengliang Xie
- Department of Urology Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen UniversityHuizhou 516200, Guangdong, China
| | - Mingjiang Dan
- Department of Urology Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen UniversityHuizhou 516200, Guangdong, China
| |
Collapse
|
6
|
Salmaninejad A, Layeghi SM, Falakian Z, Golestani S, Kobravi S, Talebi S, Yousefi M. An update to experimental and clinical aspects of tumor-associated macrophages in cancer development: hopes and pitfalls. Clin Exp Med 2024; 24:156. [PMID: 39003350 PMCID: PMC11246281 DOI: 10.1007/s10238-024-01417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant tumor-infiltrating stromal cells, and their normal function in tumor microenvironment (TME) is to suppress tumor cells by producing cytokines which trigger both direct cell cytotoxicity and antibody-mediated immune response. However, upon prolonged exposure to TME, the classical function of these so-called M1-type TAMs can be converted to another type, "M2-type," which are recruited by tumor cells so that they promote tumor growth and metastasis. This is the reason why the accumulation of TAMs in TME is correlated with poor prognosis in cancer patients. Both M1- and M2-types have high degree of plasticity, and M2-type cells can be reprogrammed to M1-type for therapeutic purposes. This characteristic introduces TAMs as promising target for developing novel cancer treatments. In addition, inhibition of M2-type cells and blocking their recruitment in TME, as well as their depletion by inducing apoptosis, are other approaches for effective immunotherapy of cancer. In this review, we summarize the potential of TAMs to be targeted for cancer immunotherapy and provide an up-to-date about novel strategies for targeting TAMs.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sepideh Mehrpour Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Falakian
- Department of Laboratory Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Shahin Golestani
- Department of Ophthalmology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Tehran Azad University, Tehran, Iran
| | - Samaneh Talebi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
7
|
Wang Y, Cheng S, Fleishman JS, Chen J, Tang H, Chen ZS, Chen W, Ding M. Targeting anoikis resistance as a strategy for cancer therapy. Drug Resist Updat 2024; 75:101099. [PMID: 38850692 DOI: 10.1016/j.drup.2024.101099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Anoikis, known as matrix detachment-induced apoptosis or detachment-induced cell death, is crucial for tissue development and homeostasis. Cancer cells develop means to evade anoikis, e.g. anoikis resistance, thereby allowing for cells to survive under anchorage-independent conditions. Uncovering the mechanisms of anoikis resistance will provide details about cancer metastasis, and potential strategies against cancer cell dissemination and metastasis. Here, we summarize the principal elements and core molecular mechanisms of anoikis and anoikis resistance. We discuss the latest progress of how anoikis and anoikis resistance are regulated in cancers. Furthermore, we summarize emerging data on selective compounds and nanomedicines, explaining how inhibiting anoikis resistance can serve as a meaningful treatment modality against cancers. Finally, we discuss the key limitations of this therapeutic paradigm and possible strategies to overcome them. In this review, we suggest that pharmacological modulation of anoikis and anoikis resistance by bioactive compounds could surmount anoikis resistance, highlighting a promising therapeutic regimen that could be used to overcome anoikis resistance in cancers.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Wenkuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Mingchao Ding
- Department of Peripheral Vascular Intervention, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China.
| |
Collapse
|
8
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
9
|
Fakhri S, Moradi SZ, Abbaszadeh F, Faraji F, Amirian R, Sinha D, McMahon EG, Bishayee A. Targeting the key players of phenotypic plasticity in cancer cells by phytochemicals. Cancer Metastasis Rev 2024; 43:261-292. [PMID: 38169011 DOI: 10.1007/s10555-023-10161-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
Plasticity of phenotypic traits refers to an organism's ability to change in response to environmental stimuli. As a result, the response may alter an organism's physiological state, morphology, behavior, and phenotype. Phenotypic plasticity in cancer cells describes the considerable ability of cancer cells to transform phenotypes through non-genetic molecular signaling activities that promote therapy evasion and tumor metastasis via amplifying cancer heterogeneity. As a result of metastable phenotypic state transitions, cancer cells can tolerate chemotherapy or develop transient adaptive resistance. Therefore, new findings have paved the road in identifying factors and agents that inhibit or suppress phenotypic plasticity. It has also investigated novel multitargeted agents that may promise new effective strategies in cancer treatment. Despite the efficiency of conventional chemotherapeutic agents, drug toxicity, development of resistance, and high-cost limit their use in cancer therapy. Recent research has shown that small molecules derived from natural sources are capable of suppressing cancer by focusing on the plasticity of phenotypic responses. This systematic, comprehensive, and critical review analyzes the current state of knowledge regarding the ability of phytocompounds to target phenotypic plasticity at both preclinical and clinical levels. Current challenges/pitfalls, limitations, and future perspectives are also discussed.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Roshanak Amirian
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, 700 026, West Bengal, India
| | - Emily G McMahon
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
10
|
Wu L, Bai L, Dai W, Wu Y, Xi P, Zhang J, Zheng L. Ginsenoside Rg3: A Review of its Anticancer Mechanisms and Potential Therapeutic Applications. Curr Top Med Chem 2024; 24:869-884. [PMID: 38441023 DOI: 10.2174/0115680266283661240226052054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Traditional Chinese Medicine (TCM) has a long history of treating various diseases and is increasingly being recognized as a complementary therapy for cancer. A promising natural compound extracted from the Chinese herb ginseng is ginsenoside Rg3, which has demonstrated significant anticancer effects. It has been tested in a variety of cancers and tumors and has proven to be effective in suppressing cancer. OBJECTIVES This work covers various aspects of the role of ginsenoside Rg3 in cancer treatment, including its biological functions, key pathways, epigenetics, and potential for combination therapies, all of which have been extensively researched and elucidated. The study aims to provide a reference for future research on ginsenoside Rg3 as an anticancer agent and a support for the potential application of ginsenoside Rg3 in cancer treatment.
Collapse
Affiliation(s)
- Lei Wu
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Bai
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenshu Dai
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontier Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaping Wu
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pengjun Xi
- Division of Infectious Diseases, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jie Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lily Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan Province, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Wang L, Zhang Y, Song Z, Liu Q, Fan D, Song X. Ginsenosides: a potential natural medicine to protect the lungs from lung cancer and inflammatory lung disease. Food Funct 2023; 14:9137-9166. [PMID: 37801293 DOI: 10.1039/d3fo02482b] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Lung cancer is the malignancy with the highest morbidity and mortality. Additionally, pulmonary inflammatory diseases, such as pneumonia, acute lung injury, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis (PF), also have high mortality rates and can promote the development and progression of lung cancer. Unfortunately, available treatments for them are limited, so it is critical to search for effective drugs and treatment strategies to protect the lungs. Ginsenosides, the main active components of ginseng, have been shown to have anti-cancer and anti-inflammatory activities. In this paper, we focus on the beneficial effects of ginsenosides on lung diseases and their molecular mechanisms. Firstly, the molecular mechanism of ginsenosides against lung cancer was summarized in detail, mainly from the points of view of proliferation, apoptosis, autophagy, angiogenesis, metastasis, drug resistance and immunity. In in vivo and in vitro lung cancer models, ginsenosides Rg3, Rh2 and CK were reported to have strong anti-lung cancer effects. Then, in the models of pneumonia and acute lung injury, the protective effect of Rb1 was particularly remarkable, followed by Rg3 and Rg1, and its molecular mechanism was mainly associated with targeting NF-κB, Nrf2, MAPK and PI3K/Akt pathways to alleviate inflammation, oxidative stress and apoptosis. Additionally, ginsenosides may also have a potential health-promoting effect in the improvement of COPD, asthma and PF. Furthermore, to overcome the low bioavailability of CK and Rh2, the development of nanoparticles, micelles, liposomes and other nanomedicine delivery systems can significantly improve the efficacy of targeted lung cancer treatment. To conclude, ginsenosides can be used as both anti-lung cancer and lung protective agents or adjuvants and have great potential for future clinical applications.
Collapse
Affiliation(s)
- Lina Wang
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Yanxin Zhang
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Zhimin Song
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Qingchao Liu
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China
- Biotechnology & Biomedicine Research Institute, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaoping Song
- Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
12
|
Shah MA, Abuzar SM, Ilyas K, Qadees I, Bilal M, Yousaf R, Kassim RMT, Rasul A, Saleem U, Alves MS, Khan H, Blundell R, Jeandet P. Ginsenosides in cancer: Targeting cell cycle arrest and apoptosis. Chem Biol Interact 2023; 382:110634. [PMID: 37451663 DOI: 10.1016/j.cbi.2023.110634] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Despite the existence of extensive clinical research and novel therapeutic treatments, cancer remains undefeated and the significant cause of death worldwide. Cancer is a disease in which growth of cells goes out of control, being also able to invade other parts of the body. Cellular division is strictly controlled by multiple checkpoints like G1/S and G2/M which, when dysregulated, lead to uncontrollable cell division. The current remedies which are being utilized to combat cancer are monoclonal antibodies, chemotherapy, cryoablation, and bone marrow transplant etc. and these have also been greatly disheartening because of their serious adverse effects like hypotension, neuropathy, necrosis, leukemia relapse and many more. Bioactive compounds derived from natural products have marked the history of the development of novel drug therapies against cancer among which ginsenosides have no peer as they target several signaling pathways, which when abnormally regulated, lead to cancer. Substantial research has reported that ginsenosides like Rb1, Rb2, Rb3, Rc, Rd, Rg3, Rh2 etc. can prevent and treat cancer by targeting different pathways and molecules by induction of autophagy, neutralizing ROS, induction of cancerous cell death by controlling the p53 pathway, modulation of miRNAs by decreasing Smad2 expression, regulating Bcl-2 expression by normalizing the NF-Kb pathway, inhibition of inflammatory pathways by decreasing the production of cytokines like IL-8, causing cell cycle arrest by restricting cyclin E1 and CDC2, and induction of apoptosis during malignancy by decreasing β-catenin levels etc. In this review, we have analyzed the anti-cancer therapeutic potential of various ginsenoside compounds in order to consider their possible use in new strategies in the fight against cancer.
Collapse
Affiliation(s)
| | - Syed Muhammad Abuzar
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Kainat Ilyas
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Irtaza Qadees
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Momna Bilal
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Rimsha Yousaf
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | | | - Azhar Rasul
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Maria Silvana Alves
- Laboratory of Cellular and Molecular Bioactivity, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Renald Blundell
- Department of Physiology and Biochemistry, Faculty of Medicine, University of Malta, Msida, MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, MSD2080 Imsida, Malta
| | - Philippe Jeandet
- University of Reims, Research Unit Induced Resistance and Plant Bioprotection USC INRAe 1488 Department of Biology and Biochemistry, Faculty of Sciences, 51100, Reims, France.
| |
Collapse
|
13
|
Janta S, Pranweerapaiboon K, Vivithanaporn P, Plubrukarn A, Chairoungdua A, Prasertsuksri P, Apisawetakan S, Chaithirayanon K. Holothurin A Inhibits RUNX1-Enhanced EMT in Metastasis Prostate Cancer via the Akt/JNK and P38 MAPK Signaling Pathway. Mar Drugs 2023; 21:345. [PMID: 37367670 DOI: 10.3390/md21060345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Due to the challenge of prostate cancer (PCa) management, there has been a surge in efforts to identify more safe and effective compounds that can modulate the epithelial-mesenchymal transition (EMT) for driving metastasis. Holothurin A (HA), a triterpenoid saponin isolated from Holothuria scabra, has now been characterized for its diverse biological activities. However, the mechanisms of HA in EMT-driven metastasis of human PCa cell lines has not yet been investigated. Moreover, runt-related transcription factor 1 (RUNX1) acts as an oncogene in prostate cancer, but little is known about its role in the EMT. Thus, the purpose of this study was to determine how RUNX1 influences EMT-mediated metastasis, as well as the potential effect of HA on EMT-mediated metastasis in endogenous and exogenous RUNX1 expressions of PCa cell lines. The results demonstrated that RUNX1 overexpression could promote the EMT phenotype with increased EMT markers, consequently driving metastatic migration and invasion in PC3 cell line through the activation of Akt/MAPK signaling pathways. Intriguingly, HA treatment could antagonize the EMT program in endogenous and exogenous RUNX1-expressing PCa cell lines. A decreasing metastasis of both HA-treated cell lines was evidenced through a downregulation of MMP2 and MMP9 via the Akt/P38/JNK-MAPK signaling pathway. Overall, our approach first demonstrated that RUNX1 enhanced EMT-driven prostate cancer metastasis and that HA was capable of inhibiting the EMT and metastatic processes and should probably be considered as a candidate for metastasis PCa treatment.
Collapse
Affiliation(s)
- Sirorat Janta
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Kanta Pranweerapaiboon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10540, Thailand
| | - Anuchit Plubrukarn
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 09112, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Somjai Apisawetakan
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Wattana, Bangkok 10110, Thailand
| | | |
Collapse
|
14
|
Nepali PR, Kyprianou N. Anoikis in phenotypic reprogramming of the prostate tumor microenvironment. Front Endocrinol (Lausanne) 2023; 14:1160267. [PMID: 37091854 PMCID: PMC10113530 DOI: 10.3389/fendo.2023.1160267] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/09/2023] [Indexed: 04/25/2023] Open
Abstract
Prostate cancer is one of the most common malignancies in males wherein 1 in 8 men are diagnosed with this disease in their lifetime. The urgency to find novel therapeutic interventions is associated with high treatment resistance and mortality rates associated with castration-resistant prostate cancer. Anoikis is an apoptotic phenomenon for normal epithelial or endothelial cells that have lost their attachment to the extracellular matrix (ECM). Tumor cells that lose their connection to the ECM can die via apoptosis or survive via anoikis resistance and thus escaping to distant organs for metastatic progression. This review discusses the recent advances made in our understanding of the signaling effectors of anoikis in prostate cancer and the approaches to translate these mechanistic insights into therapeutic benefits for reducing lethal disease outcomes (by overcoming anoikis resistance). The prostate tumor microenvironment is a highly dynamic landscape wherein the balance between androgen signaling, cell lineage changes, epithelial-mesenchymal transition (EMT), extracellular matrix interactions, actin cytoskeleton remodeling as well as metabolic changes, confer anoikis resistance and metastatic spread. Thus, these mechanisms also offer unique molecular treatment signatures, exploitation of which can prime prostate tumors to anoikis induction with a high translational significance.
Collapse
Affiliation(s)
- Prerna R. Nepali
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Natasha Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
15
|
Jung DH, Nahar J, Mathiyalagan R, Rupa EJ, Ramadhania ZM, Han Y, Yang DC, Kang SC. A Focused Review on Molecular Signalling Mechanisms of Ginsenosides Anti-Lung Cancer and Anti-inflammatory Activities. Anticancer Agents Med Chem 2023; 23:3-14. [PMID: 35319393 DOI: 10.2174/1871520622666220321091022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/01/2021] [Accepted: 12/12/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ginseng (Panax ginseng Meyer) is a cultivated medicinal herb that has been widely available in the Asian region since the last century. Ginseng root is used worldwide in Oriental medicine. Currently, the global mortality and infection rates for lung cancer and inflammation are significantly increasing. Therefore, various preventative methods related to the activity of ginsenosides have been used for lung cancer as well as inflammation. METHODS Web-based searches were performed on Web of Science, Springer, PubMed, and Scopus. A cancer statistical analysis was also conducted to show the current ratio of affected cases and death from lung cancer around the world. RESULTS Ginsenosides regulate the enzymes that participate in tumor growth and migration, such as nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (p38 MAPK), c-Jun N-terminal kinase (JNK), extracellular signalregulated kinases 1/2 (ERK1/2), the gelatinase network metalloproteinase-2 (MMP-2/9) and activator protein 1 (AP-1). In addition, ginsenosides also possess anti-inflammatory effects by inhibiting the formation of proinflammatory cytokines (tumor necrosis factor-α) (TNF-α) and interleukin-1β (IL-1β) and controlling the activities of inflammatory signalling pathways, such as NF-κB, Janus kinase2/signal transducer, and activator of transcription 3 (Jak2/Stat3). CONCLUSION In several in vitro and in vivo models, P. ginseng showed potential beneficial effects in lung cancer and inflammation treatment. In this review, we provide a detailed and up-to-date summary of research evidence for antilung cancer and anti-inflammatory protective effects of ginsenosides and their potential molecular mechanisms.
Collapse
Affiliation(s)
- Dae-Hyo Jung
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Jinnatun Nahar
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Esrat Jahan Rupa
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Zelika Mega Ramadhania
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Yaxi Han
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Deok-Chun Yang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.,Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Se Chan Kang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| |
Collapse
|
16
|
Recent advances in ginsenosides against respiratory diseases: Therapeutic targets and potential mechanisms. Biomed Pharmacother 2023; 158:114096. [PMID: 36502752 DOI: 10.1016/j.biopha.2022.114096] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Respiratory diseases mainly include asthma, influenza, pneumonia, chronic obstructive pulmonary disease, pulmonary hypertension, lung fibrosis, and lung cancer. Given their high prevalence and poor prognosis, the prevention and treatment of respiratory diseases are increasingly essential. In particular, the development for the novel strategies of drug treatment has been a hot topic in the research field. Ginsenosides are the major component of Panax ginseng C. A. Meyer (ginseng), a food homology and well-known medicinal herb. In this review, we summarize the current therapeutic effects and molecular mechanisms of ginsenosides in respiratory diseases. METHODS The reviewed studies were retrieved via a thorough analysis of numerous articles using electronic search tools including Sci-Finder, ScienceDirect, PubMed, and Web of Science. The following keywords were used for the online search: ginsenosides, asthma, influenza, pneumonia, chronic obstructive pulmonary disease (COPD), pulmonary hypertension (PH), lung fibrosis, lung cancer, and clinical trials. We summarized the findings and the conclusions from 176 manuscripts on ginsenosides, including research articles and reviews. RESULTS Ginsenosides Rb1, Rg1, Rg3, Rh2, and CK, which are the most commonly reported ginsenosides for treating of respiratory diseases, and other ginsenosides such as Rh1, Rk1, Rg5, Rd and Re, all primarily reduce pneumonia, fibrosis, and inhibit tumor progression by targeting NF-κB, TGF-β/Smad, PI3K/AKT/mTOR, and JNK pathways, thereby ameliorating respiratory diseases. CONCLUSION This review provides novel ideas and important aspects for the future research of ginsenosides for treating respiratory diseases.
Collapse
|
17
|
Ye XW, Li CS, Zhang HX, Li Q, Cheng SQ, Wen J, Wang X, Ren HM, Xia LJ, Wang XX, Xu XF, Li XR. Saponins of ginseng products: a review of their transformation in processing. Front Pharmacol 2023; 14:1177819. [PMID: 37188270 PMCID: PMC10175582 DOI: 10.3389/fphar.2023.1177819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The primary processed product of Panax ginseng C.A. Meyer (P. ginseng) is red ginseng. As technology advances, new products of red ginseng have arisen. Red ginseng products, e.g., traditional red ginseng, sun ginseng, black ginseng, fermented red ginseng, and puffed red ginseng, are commonly used in herbal medicine. Ginsenosides are the major secondary metabolites of P. ginseng. The constituents of P. ginseng are significantly changed during processing, and several pharmacological activities of red ginseng products are dramatically increased compared to white ginseng. In this paper, we aimed to review the ginsenosides and pharmacological activities of various red ginseng products, the transformation law of ginsenosides in processing, and some clinical trials of red ginseng products. This article will help to highlight the diverse pharmacological properties of red ginseng products and aid in the future development of red ginseng industrialization.
Collapse
Affiliation(s)
- Xian-Wen Ye
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun-Shuai Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Hai-Xia Zhang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Shui-Qing Cheng
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Jia Wen
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Xuan Wang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Hong-Min Ren
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Liang-Jing Xia
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Xu-Xing Wang
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Fang Xu
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Xin-Fang Xu, ; Xiang-Ri Li,
| | - Xiang-Ri Li
- Centre of TCM Processing Research, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Xin-Fang Xu, ; Xiang-Ri Li,
| |
Collapse
|
18
|
Wang J, Luo Z, Lin L, Sui X, Yu L, Xu C, Zhang R, Zhao Z, Zhu Q, An B, Wang Q, Chen B, Leung ELH, Wu Q. Anoikis-Associated Lung Cancer Metastasis: Mechanisms and Therapies. Cancers (Basel) 2022; 14:cancers14194791. [PMID: 36230714 PMCID: PMC9564242 DOI: 10.3390/cancers14194791] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 12/08/2022] Open
Abstract
Simple Summary Anoikis is a programmed cell death process resulting from the loss of interaction between cells and the extracellular matrix. Therefore, it is necessary to overcome anoikis when tumor cells acquire metastatic potential. In lung cancer, the composition of the extracellular matrix, cell adhesion-related membrane proteins, cytoskeletal regulators, and epithelial–mesenchymal transition are involved in the process of anoikis, and the initiation of apoptosis signals is a critical step in anoikis. Inversely, activation of growth signals counteracts anoikis. This review summarizes the regulators of lung cancer-related anoikis and explores potential drug applications targeting anoikis. Abstract Tumor metastasis occurs in lung cancer, resulting in tumor progression and therapy failure. Anoikis is a mechanism of apoptosis that combats tumor metastasis; it inhibits the escape of tumor cells from the native extracellular matrix to other organs. Deciphering the regulators and mechanisms of anoikis in cancer metastasis is urgently needed to treat lung cancer. Several natural and synthetic products exhibit the pro-anoikis potential in lung cancer cells and in vivo models. These products include artonin E, imperatorin, oroxylin A, lupalbigenin, sulforaphane, renieramycin M, avicequinone B, and carbenoxolone. This review summarizes the current understanding of the molecular mechanisms of anoikis regulation and relevant regulators involved in lung cancer metastasis and discusses the therapeutic potential of targeting anoikis in the treatment of lung cancer metastasis.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Zhijie Luo
- The First Clinical Medical College, The First Hospital Affiliated, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Lizhu Lin
- The First Clinical Medical College, The First Hospital Affiliated, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xinbing Sui
- School of Pharmacy, Department of Medical Oncology, Hangzhou Normal University, Hangzhou 311121, China
| | - Lili Yu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Cong Xu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Ruonan Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Ziming Zhao
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qianru Zhu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Bo An
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qiao Wang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Bi Chen
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau 999078, China
- Correspondence: (E.L.-H.L.); (Q.W.)
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangdong University of Technology, Guangzhou 510006, China
- Zhuhai MUST Science and Technology Research Institute, Zhuhai 519031, China
- Correspondence: (E.L.-H.L.); (Q.W.)
| |
Collapse
|
19
|
Oh HM, Cho CK, Son CG. Experimental Evidence for the Anti-Metastatic Action of Ginsenoside Rg3: A Systematic Review. Int J Mol Sci 2022; 23:9077. [PMID: 36012338 PMCID: PMC9409359 DOI: 10.3390/ijms23169077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/26/2022] Open
Abstract
Cancer metastasis is the leading cause of death in cancer patients. Due to the limitations of conventional cancer treatment, such as chemotherapy, there is a need for novel therapeutics to prevent metastasis. Ginsenoside Rg3, a major active component of Panax ginseng C.A. Meyer, inhibits tumor growth and has the potential to prevent tumor metastasis. Herein, we systematically reviewed the anti-metastatic effects of Rg3 from experimental studies. We searched for articles in three research databases, MEDLINE (PubMed), EMBASE, and the Cochrane Central Register of Controlled Trials (CENTRAL) through March 2022. In total, 14 studies (eight animal and six in vitro) provide data on the anti-metastatic effects of Rg3 and the relevant mechanisms. The major anti-metastatic mechanisms of Rg3 involve cancer stemness, epithelial mesenchymal transition (EMT) behavior, and angiogenesis. Taken together, Rg3 would be one of the herbal resources in anti-metastatic drug developments through further well-designed investigations and clinical studies. Our review provides valuable reference data for Rg3-derived studies targeting tumor metastasis.
Collapse
Affiliation(s)
- Hyeon-Muk Oh
- College of Korean Medicine, Daejeon University, Daejeon 35235, Korea
| | - Chong-Kwan Cho
- College of Korean Medicine, Daejeon University, Daejeon 35235, Korea
- East-West Cancer Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon 35235, Korea
| | - Chang-Gue Son
- College of Korean Medicine, Daejeon University, Daejeon 35235, Korea
- Liver and Immunology Research Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon 35235, Korea
| |
Collapse
|
20
|
Mao Y, Meng L, Liu H, Lu Y, Yang K, Ouyang G, Ban Y, Chen S. Therapeutic potential of traditional Chinese medicine for vascular endothelial growth factor. J Zhejiang Univ Sci B 2022; 23:353-364. [PMID: 35557037 DOI: 10.1631/jzus.b2101055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vascular endothelial growth factor (VEGF) is the main regulator of physiological angiogenesis during embryonic development, bone growth, and reproductive function, and it also participates in a series of pathological changes. Traditional Chinese medicine (TCM), with a history of more than 2000 years, has been widely used in clinical practice, while the exploration of its mechanisms has only begun. This review summarizes the research of recent years on the influence of TCM on VEGF. It is found that many Chinese medicines and recipes have a regulatory effect on VEGF, indicating that Chinese medicine has broad prospects as a complementary and alternative therapy, providing new treatment ideas for clinical applications and the theoretical basis for research on the mechanisms of TCM.
Collapse
Affiliation(s)
- Yijia Mao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Lingkai Meng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Huayi Liu
- Department of Digestive Diseases, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China.
| | - Yuting Lu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Kuo Yang
- Department of Digestive Diseases, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Guangze Ouyang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Yanran Ban
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| | - Shuang Chen
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301608, China
| |
Collapse
|
21
|
Ge H, Xu C, Chen H, Liu L, Zhang L, Wu C, Lu Y, Yao Q. Traditional Chinese Medicines as Effective Reversals of Epithelial-Mesenchymal Transition Induced-Metastasis of Colorectal Cancer: Molecular Targets and Mechanisms. Front Pharmacol 2022; 13:842295. [PMID: 35308223 PMCID: PMC8931761 DOI: 10.3389/fphar.2022.842295] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer worldwide. Distant metastasis is the major cause of cancer-related mortality in patients with CRC. Epithelial-mesenchymal transition (EMT) is a critical process triggered during tumor metastasis, which is also the main impetus and the essential access within this duration. Therefore, targeting EMT-related molecular pathways has been considered a novel strategy to explore effective therapeutic agents against metastatic CRC. Traditional Chinese medicines (TCMs) with unique properties multi-target and multi-link that exert their therapeutic efficacies holistically, which could inhibit the invasion and metastasis ability of CRC cells via inhibiting the EMT process by down-regulating transforming growth factor-β (TGF-β)/Smads, PI3K/Akt, NF-κB, Wnt/β-catenin, and Notch signaling pathways. The objective of this review is to summarize and assess the anti-metastatic effect of TCM-originated bioactive compounds and Chinese medicine formulas by mediating EMT-associated signaling pathways in CRC therapy, providing a foundation for further research on the exact mechanisms of action through which TCMs affect EMT transform in CRC.
Collapse
Affiliation(s)
- Hongzhang Ge
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Chao Xu
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Haitao Chen
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ling Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Changhong Wu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Lu
- Department of Clinical Nutrition, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qinghua Yao
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Clinical Nutrition, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: Qinghua Yao,
| |
Collapse
|
22
|
LI H, WANG T, CUI W, GAO Z, CHE Z. Effect of ginsenoside Rg3 on proliferation and apoptosis of 786-0 cells and AktmTORSTAT3 signaling in renal carcinoma. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.124121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
| | | | - Wei CUI
- Zibo Central Hospital, China
| | | | - Zi CHE
- Zibo Central Hospital, China
| |
Collapse
|
23
|
Ayanlaja AA, Hong X, Cheng B, Zhou H, Kanwore K, Alphayo-Kambey P, Zhang L, Tang C, Adeyanju MM, Gao D. Susceptibility of cytoskeletal-associated proteins for tumor progression. Cell Mol Life Sci 2021; 79:13. [PMID: 34964908 PMCID: PMC11072373 DOI: 10.1007/s00018-021-04101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 10/19/2022]
Abstract
The traditional functions of cytoskeletal-associated proteins (CAPs) in line with polymerization and stabilization of the cytoskeleton have evolved and are currently underrated in oncology. Although therapeutic drugs have been developed to target the cytoskeletal components directly in cancer treatment, several recently established therapeutic agents designed for new targets block the proliferation of cancer cells and suppress resistance to existing target agents. It would seem like these targets only work toward inhibiting the polymerization of cytoskeletal components or hindering mitotic spindle formation in cancer cells, but a large body of literature points to CAPs and their culpability in cell signaling, molecular conformation, organelle trafficking, cellular metabolism, and genomic modifications. Here, we review those underappreciated functions of CAPs, and we delineate the implications of cellular signaling instigated by evasive properties induced by aberrant expression of CAPs in response to stress or failure to exert normal functions. We present an analogy establishing CAPs as vulnerable targets for cancer systems and credible oncotargets. This review establishes a paradigm in which the cancer machinery may commandeer the conventional functions of CAPs for survival, drug resistance, and energy generation; an interesting feature overdue for attention.
Collapse
Affiliation(s)
- Abiola Abdulrahman Ayanlaja
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Neurology, Johns Hopkins University School of Medicine, 201 N Broadway, Baltimore, MD, 21287, USA
| | - Xiaoliang Hong
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bo Cheng
- The Affiliated Oriental Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Han Zhou
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kouminin Kanwore
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Piniel Alphayo-Kambey
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Lin Zhang
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chuanxi Tang
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | | | - Dianshuai Gao
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
24
|
Zhou Y, Farooqi AA, Xu B. Comprehensive review on signaling pathways of dietary saponins in cancer cells suppression. Crit Rev Food Sci Nutr 2021:1-26. [PMID: 34751072 DOI: 10.1080/10408398.2021.2000933] [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] [Indexed: 01/10/2023]
Abstract
Nutrigenomics utilizes high-throughput genomic technologies to reveal changes in gene and protein levels. Excitingly, ever-growing body of scientific findings has provided sufficient evidence about the interplay between diet and genes. Cutting-edge research and advancements in genomics, epigenetics and metabolomics have deepened our understanding on the role of dietary factors in the inhibition of carcinogenesis and metastasis. Dietary saponins, a type of triterpene glycosides, are generally found in Platycodon grandifloras, Dioscorea oppositifolia, asparagus, legumes, and sea cucumber. Wealth of information has started to shed light on pleiotropic mechanistic roles of dietary saponins in cancer prevention and inhibition. In this review, we have attempted to summarize the in vitro research of dietary saponins in the last two decades by searching common databases such as Google Scholar, PubMed, Scopus, and Web of Science. The results showed that dietary saponins exerted anti-cancer activities via regulation of apoptosis, autophagy, arrest cell cycle, anti-proliferation, anti-metastasis, and anti-angiogenesis, by regulation of several critical signaling pathways, including MAPK, PI3K/Akt/mTOR, NF-κB, and VEGF/VEGFR. However, there is no data about the dosage of dietary saponins for practical anti-cancer effects in human bodies. Extensive clinical studies are needed to confirm the effectiveness of dietary saponins for further commercial and medical applications.
Collapse
Affiliation(s)
- Yifan Zhou
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, Guangdong, China.,Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| | | | - Baojun Xu
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, Guangdong, China
| |
Collapse
|
25
|
Cai Z, Teng Y, Chen Y. The Effect of Shenyi Capsule on Non-Small-Cell Lung Cancer Combined with Chemotherapy from the Yin-Yang Perspective. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:1653750. [PMID: 34512772 PMCID: PMC8426066 DOI: 10.1155/2021/1653750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/26/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
As an example of Shenyi capsule on non-small-cell lung cancer combined with chemotherapy, this review discusses the synergistic effect and mechanism of natural drugs in oncotherapy from the yin-yang perspective in ancient Chinese philosophy, so as to reflect the therapeutic principle of natural drugs for tumor more comprehensively. The major focuses of this review are on the philosophical thinking of yin-yang as a tool which can not only explain the effect of Shenyi capsule in NSCLC combined with chemotherapy but also explore the mechanism of Shenyi capsule at the cellular and molecular level. Learning from the "yin-yang" thinking of ancient Chinese philosophy will bring more enlightenment to the research and development of traditional Chinese drugs in the future.
Collapse
Affiliation(s)
- Zhixing Cai
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Yue Teng
- Outpatient Department of Clinic Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai 200437, China
| | - Yue Chen
- Department of T.C.M, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| |
Collapse
|
26
|
ADAM 17 and Epithelial-to-Mesenchymal Transition: The Evolving Story and Its Link to Fibrosis and Cancer. J Clin Med 2021; 10:jcm10153373. [PMID: 34362154 PMCID: PMC8347979 DOI: 10.3390/jcm10153373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
For decades, metalloproteinase 17 (ADAM17) has been the goal of wide investigation. Since its discovery as the tumour necrosis factor-α convertase, it has been studied as the main drug target, especially in the context of inflammatory conditions and tumour. In fact, evidence is mounting to support a key role of ADAM17 in the induction of the proliferation, migration and progression of tumour cells and the trigger of the pro-fibrotic process during chronic inflammatory conditions; this occurs, probably, through the activation of epithelial-to-mesenchymal transition (EMT). EMT is a central morphologic conversion that occurs in adults during wound healing, tumour progression and organ fibrosis. EMT is characterised by the disassembly of cell–cell contacts, remodelling of the actin cytoskeleton and separation of cells, and generates fibroblast-like cells that express mesenchymal markers and have migratory properties. This transition is characterised by loss of epithelial proteins such as E-cadherin and the acquisition of new mesenchymal markers, including vimentin and a-smooth muscle actin. The present review discusses the current understanding of molecular mechanisms involved in ADAM17-dependent EMT in order to individuate innovative therapeutic strategies using ADAM17-related pathways.
Collapse
|
27
|
Nakhjavani M, Smith E, Yeo K, Palethorpe HM, Tomita Y, Price TJ, Townsend AR, Hardingham JE. Anti-Angiogenic Properties of Ginsenoside Rg3 Epimers: In Vitro Assessment of Single and Combination Treatments. Cancers (Basel) 2021; 13:cancers13092223. [PMID: 34066403 PMCID: PMC8125638 DOI: 10.3390/cancers13092223] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
Tumour angiogenesis plays a key role in tumour growth and progression. The application of current anti-angiogenic drugs is accompanied by adverse effects and drug resistance. Therefore, finding safer effective treatments is needed. Ginsenoside Rg3 (Rg3) has two epimers, 20(S)-Rg3 (SRg3) and 20(R)-Rg3 (RRg3), with stereoselective activities. Using response surface methodology, we optimised a combination of these two epimers for the loop formation of human umbilical vein endothelial cell (HUVEC). The optimised combination (C3) was tested on HUVEC and two murine endothelial cell lines. C3 significantly inhibited the loop formation, migration, and proliferation of these cells, inducing apoptosis in HUVEC and cell cycle arrest in all of the cell lines tested. Using molecular docking and vascular endothelial growth factor (VEGF) bioassay, we showed that Rg3 has an allosteric modulatory effect on vascular endothelial growth factor receptor 2 (VEGFR2). C3 also decreased the VEGF expression in hypoxic conditions, decreased the expression of aquaporin 1 and affected AKT signaling. The proteins that were mostly affected after C3 treatment were those related to mammalian target of rapamycin (mTOR). Eukaryotic translation initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) was one of the important targets of C3, which was affected in both hypoxic and normoxic conditions. In conclusion, these results show the potential of C3 as a novel anti-angiogenic drug.
Collapse
Affiliation(s)
- Maryam Nakhjavani
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (K.Y.); (Y.T.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
| | - Eric Smith
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (K.Y.); (Y.T.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Correspondence: ; Tel.: +61-8-8222-6142
| | - Kenny Yeo
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (K.Y.); (Y.T.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
| | - Helen M. Palethorpe
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia;
| | - Yoko Tomita
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (K.Y.); (Y.T.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Tim J. Price
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Amanda R. Townsend
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Jennifer E. Hardingham
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (M.N.); (K.Y.); (Y.T.); (J.E.H.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; (T.J.P.); (A.R.T.)
| |
Collapse
|
28
|
Liu Z, Liu T, Li W, Li J, Wang C, Zhang K. Insights into the antitumor mechanism of ginsenosides Rg3. Mol Biol Rep 2021; 48:2639-2652. [PMID: 33661439 DOI: 10.1007/s11033-021-06187-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/28/2021] [Indexed: 12/12/2022]
Abstract
Panax ginseng, an ancient herb, belonging to Chinese traditional medicine, is an important herb that has a remarkable impact on various diseases. Ginsenoside Rg3, one of the most abundant ginsenosides, exerts significant functions in the prevention of various types of cancers with few side effects. In the present review, its functional molecular mechanisms are explored, including the improvement of antioxidant and anti-inflammation properties, immune regulation, induction of tumor apoptosis, prevention of tumor invasion and metastasis, tumor proliferation and angiogenesis, and reduction of chemoresistance and radioresistance. On the other hand, metabolism, pharmacokinetics and clinical indications of Rg3 are also discussed. The biological functional role of ginsenoside Rg3 may be associated with that it is a steroid glycoside with diverse biological activities and many signaling pathway can be regulated. Many clinical trials are highly needed to confirm the functions of ginsenoside Rg3.
Collapse
Affiliation(s)
- Zongyu Liu
- Department of General Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China
| | - Wei Li
- Department of General Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China
| | - Cuizhu Wang
- Department of New Drug Research Office, College of Pharmacy of Jilin University, Changchun, 130000, China
| | - Kai Zhang
- Department of General Surgery, The Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130000, China.
| |
Collapse
|
29
|
Anti-Metastatic and Anti-Inflammatory Effects of Matrix Metalloproteinase Inhibition by Ginsenosides. Biomedicines 2021; 9:biomedicines9020198. [PMID: 33671187 PMCID: PMC7921986 DOI: 10.3390/biomedicines9020198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes which cleave extracellular matrix (ECM) and other substrates. They are deeply involved in both cancer metastasis and human chronic inflammatory diseases such as osteoarthritis and Crohn’s disease. Regulation of MMPs is closely associated with signaling molecules, especially mitogen-activated protein kinases (MAPKs), including three representative kinases, extracellular signal regulated kinases (ERK), p38 and c-Jun N-terminal kinases (JNK). Ginseng (Panax sp.) is a plant which has been traditionally used for medicinal applications. Ginsenosides are major metabolites which have potentials to treat various human diseases. In this review, the pharmacological effects of ginsenosides have been rigorously investigated; these include anti-metastatic and anti-inflammatory activities of ginsenosides associated with suppression of MMPs via regulation of various signaling pathways. This will highlight the importance of MMPs as therapeutic targets for anti-metastatic and anti-inflammatory drug development based on ginsenosides.
Collapse
|
30
|
Hong S, Cai W, Huang Z, Wang Y, Mi X, Huang Y, Lin Z, Chen X. Ginsenoside Rg3 enhances the anticancer effect of 5‑FU in colon cancer cells via the PI3K/AKT pathway. Oncol Rep 2020; 44:1333-1342. [PMID: 32945504 PMCID: PMC7448421 DOI: 10.3892/or.2020.7728] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy is one of the most commonly used treatments for patients with advanced colon cancer, yet the toxicity of chemotherapy agents, such as 5‑fluorouracil (5‑FU), limits the effectiveness of chemotherapy. Ginsenoside Rg3 (Rg3) is an active ingredient isolated from ginseng. Rg3 has been shown to display anticancer effects on a variety of malignancies. Yet, whether Rg3 synergizes the effect of 5‑FU to inhibit the growth of human colon cancer remains unknown. The present study was designed to ascertain whether Rg3 is able to enhance the anti‑colon cancer effect of 5‑FU. The results revealed that combined treatment of Rg3 and 5‑FU significantly enhanced the inhibition of the proliferation, colony formation, invasion and migration of human colon cancer cells (SW620 and LOVO) in vitro. We also found that combined treatment of Rg3 and 5‑FU significantly enhanced the apoptosis of colon cancer cells by activating the Apaf1/caspase 9/caspase 3 pathway and arrested the cell cycle of the colon cancer cells in G0/G1 by promoting the expression of Cyclin D1, CDK2 and CDK4. In addition, the PI3K/AKT signaling pathway in colon cancer cells was suppressed by Rg3 and 5‑FU. In vivo, Rg3 synergized the effect of 5‑FU to inhibit the growth of human colon cancer xenografts in nude mice. Similarly, combined treatment of Rg3 and 5‑FU altered the expression of colon cancer protein in vivo and in vitro. Collectively, the present study demonstrated that ginsenoside Rg3 enhances the anticancer effect of 5‑FU in colon cancer cells via the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Shunzhong Hong
- Endoscopy Center, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Wenjie Cai
- Department of Tumor Radiotherapy, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Zicheng Huang
- Department of Gastroenterology, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Yubin Wang
- Department of Gastroenterology, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Xifeng Mi
- Endoscopy Center, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Yisen Huang
- Department of Gastroenterology, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Zhijin Lin
- Department of Gastroenterology, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Xiangbo Chen
- Endoscopy Center, The Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| |
Collapse
|
31
|
Study on the Mechanism of Ginseng in the Treatment of Lung Adenocarcinoma Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2658795. [PMID: 32802118 PMCID: PMC7415121 DOI: 10.1155/2020/2658795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/12/2020] [Indexed: 02/03/2023]
Abstract
Background Ginseng, a traditional Chinese medicine, was used to prevent and treat many diseases such as diabetes, inflammation, and cancer. In recent years, there are some reports about the treatment of lung adenocarcinoma with ginseng monomer compounds, but there is no systematic study on the related core targets and mechanism of ginseng in the treatment of lung adenocarcinoma up to now. Therefore, this study systematically and comprehensively studied the molecular mechanism of ginseng in the treatment of lung adenocarcinoma based on network pharmacology and further proved the potential targets by A549 cell experiments for the first time. Methods The targets of disease and drug were obtained from Gene database. Subsequently, the compound-target network was constructed, and the core potential targets were screened out by plug-in into Cytoscape. Furthermore, the core targets and mechanism of ginseng in the treatment of lung adenocarcinoma were verified by MTT test, cell scratch test, immunohistochemistry, and qRT-PCR. Results 1791 disease targets and 144 drug targets were obtained by searching the Gene database. Meanwhile, 15 core targets were screened out: JUN, MAPK8, PTGS2, CASP3, VEGFA, MMP9, AKT1, TNF, FN1, FOS, MMP782, IL-1β, IL-2, ICAM1, and HMOX1. The results of cell experiments indicate that ginseng could treat lung adenocarcinoma by cell proliferation, migration, and apoptosis. In addition, according to the results of the 15 core targets by qRT-PCR, JUN, IL-1β, IL-2, ICAM1, HMOX1, MMP9, and MMP2 are upregulated core targets, while PTGS2 and TNF are downregulated core targets. Conclusion This study systematically and comprehensively studied 15 core targets by network pharmacology for the first time. Subsequently, it is verified that 9 core targets for ginseng treatment of lung adenocarcinoma, namely, JUN, IL-1β, IL-2, ICAM1, HMOX1, MMP9, MMP2, PTGS2, and TNF, are closely related to the proliferation, migration, and apoptosis of lung adenocarcinoma cells. This study has reference value for the clinical application of ginseng in the treatment of lung adenocarcinoma.
Collapse
|
32
|
Kim H, Choi P, Kim T, Kim Y, Song BG, Park YT, Choi SJ, Yoon CH, Lim WC, Ko H, Ham J. Ginsenosides Rk1 and Rg5 inhibit transforming growth factor-β1-induced epithelial-mesenchymal transition and suppress migration, invasion, anoikis resistance, and development of stem-like features in lung cancer. J Ginseng Res 2020; 45:134-148. [PMID: 33437165 PMCID: PMC7790904 DOI: 10.1016/j.jgr.2020.02.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/03/2020] [Accepted: 02/28/2020] [Indexed: 01/10/2023] Open
Abstract
Background Lung cancer has a high incidence worldwide, and most lung cancer-associated deaths are attributable to cancer metastasis. Although several medicinal properties of Panax ginseng Meyer have been reported, the effect of ginsenosides Rk1 and Rg5 on epithelial-mesenchymal transition (EMT) stimulated by transforming growth factor beta 1 (TGF- β1) and self-renewal in A549 cells is relatively unknown. Methods We treated TGF-β1 or alternatively Rk1 and Rg5 in A549 cells. We used western blot analysis, real-time polymerase chain reaction (qPCR), wound healing assay, Matrigel invasion assay, and anoikis assays to determine the effect of Rk1 and Rg5 on TGF-mediated EMT in lung cancer cell. In addition, we performed tumorsphere formation assays and real-time PCR to evaluate the stem-like properties. Results EMT is induced by TGF-β1 in A549 cells causing the development of cancer stem-like features. Expression of E-cadherin, an epithelial marker, decreased and an increase in vimentin expression was noted. Cell mobility, invasiveness, and anoikis resistance were enhanced with TGF-β1 treatment. In addition, the expression of stem cell markers, CD44, and CD133, was also increased. Treatment with Rk1 and Rg5 suppressed EMT by TGF-β1 and the development of stemness in a dose-dependent manner. Additionally, Rk1 and Rg5 markedly suppressed TGF-β1-induced metalloproteinase-2/9 (MMP2/9) activity, and activation of Smad2/3 and nuclear factor kappa B/extra-cellular signal regulated kinases (NF-kB/ERK) pathways in lung cancer cells. Conclusions Rk1 and Rg5 regulate the EMT inducing TGF-β1 by suppressing the Smad and NF-κB/ERK pathways (non-Smad pathway).
Collapse
Affiliation(s)
- Hyunhee Kim
- Department of Biomedical Sciences, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Pilju Choi
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Taejung Kim
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Youngseok Kim
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Bong Geun Song
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Young-Tae Park
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Seon-Jun Choi
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Cheol Hee Yoon
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
| | - Won-Chul Lim
- Traditional Food Research Group, Korea Food Research Institute, Wanju, Republic of Korea
| | - Hyeonseok Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Republic of Korea
- Corresponding author. Biomedical Research Center, Asan Institute for Life Sciences, Seoul 05505, Republic of Korea
| | - Jungyeob Ham
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, Republic of Korea
- Corresponding author. Natural Products Research Institute, Korea Institute of Science and Technology (KIST), 679 Saimdang-ro, Gangneung, 25451, Republic of Korea.
| |
Collapse
|
33
|
Strouhalova K, Přechová M, Gandalovičová A, Brábek J, Gregor M, Rosel D. Vimentin Intermediate Filaments as Potential Target for Cancer Treatment. Cancers (Basel) 2020; 12:E184. [PMID: 31940801 PMCID: PMC7017239 DOI: 10.3390/cancers12010184] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Intermediate filaments constitute the third component of the cellular skeleton. Unlike actin and microtubule cytoskeletons, the intermediate filaments are composed of a wide variety of structurally related proteins showing distinct expression patterns in tissues and cell types. Changes in the expression patterns of intermediate filaments are often associated with cancer progression; in particular with phenotypes leading to increased cellular migration and invasion. In this review we will describe the role of vimentin intermediate filaments in cancer cell migration, cell adhesion structures, and metastasis formation. The potential for targeting vimentin in cancer treatment and the development of drugs targeting vimentin will be reviewed.
Collapse
Affiliation(s)
- Katerina Strouhalova
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Magdalena Přechová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Aneta Gandalovičová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Daniel Rosel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| |
Collapse
|
34
|
Chen H, Yang H, Fan D, Deng J. The Anticancer Activity and Mechanisms of Ginsenosides: An Updated Review. EFOOD 2020. [DOI: 10.2991/efood.k.200512.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
35
|
Wang C, Liu J, Deng J, Wang J, Weng W, Chu H, Meng Q. Advances in the chemistry, pharmacological diversity, and metabolism of 20( R)-ginseng saponins. J Ginseng Res 2020; 44:14-23. [PMID: 32095093 PMCID: PMC7033361 DOI: 10.1016/j.jgr.2019.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 11/14/2018] [Accepted: 01/21/2019] [Indexed: 12/15/2022] Open
Abstract
Ginseng has been used as a popular herbal medicine in East Asia for at least two millennia. However, 20(R)-ginseng saponins, one class of important rare ginsenosides, are rare in natural products. 20(R)-ginseng saponins are generally prepared by chemical epimerization and microbial transformation from 20(S)-isomers. The C20 configuration of 20(R)-ginseng saponins are usually determined by 13C NMR and X-ray single-crystal diffraction. 20(R)-ginseng saponins have antitumor, antioxidative, antifatigue, neuroprotective, and osteoclastogenesis inhibitory effects, among others. Owing to the chemical structure and pharmacological and stereoselective properties, 20(R)-ginseng saponins have attracted a great deal of attention in recent years. In this study, the discovery, identification, chemical epimerization, microbial transformation, pharmacological activities, and metabolism of 20(R)-ginseng saponins are summarized.
Collapse
Affiliation(s)
- Chaoming Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Juan Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jianqiang Deng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jiazhen Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Weizhao Weng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Hongxia Chu
- Department of Cardiovascular Medicine, Yuhuangding Hospital of Yantai, Shandong, China
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
36
|
Phi LTH, Wijaya YT, Sari IN, Kim KS, Yang YG, Lee MW, Kwon HY. 20(R)-Ginsenoside Rg3 Influences Cancer Stem Cell Properties and the Epithelial-Mesenchymal Transition in Colorectal Cancer via the SNAIL Signaling Axis. Onco Targets Ther 2019; 12:10885-10895. [PMID: 31849492 PMCID: PMC6912006 DOI: 10.2147/ott.s219063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Cancer stem cells (CSCs) have been proposed as central drivers of cancer relapse in many cancers. In the present study, we investigated the inhibitory effect of 20(R)-Ginsenoside Rg3 (Rg3R), a major active component of ginseng saponin, on CSC-like cells and the Epithelial-Mesenchymal Transition (EMT) in colorectal cancer (CRC). Methods The effects of ginsenoside Rg3R on the colony-forming, migration, invasion, and wound-healing abilities of CRC cells were determined in HT29 and SW620 cell lines in vitro. Further, ginsenoside Rg3R was given intraperitoneally at 5mg/kg of mouse body weight to check its effect on the metastasis of CRC cells in vivo. Results Ginsenoside Rg3R significantly inhibited CSC properties, but did not affect cell proliferation. Moreover, ginsenoside Rg3R treatment significantly inhibited the motility of CRC cells based on migration, invasion, and wound-healing assays. The inhibitory effects of ginsenoside Rg3R on CRC are potentially mediated by significant down-regulation of the expression of stemness genes and EMT markers in CRC cells in a SNAIL-dependent manner. Furthermore, ginsenoside Rg3R treatment decreased both the number and size of tumor nodules in the liver, lung, and kidney tissues in a metastasis mouse model. Conclusion These findings highlighted the potential use of ginsenoside Rg3R in clinical applications for colorectal cancer treatment.
Collapse
Affiliation(s)
- Lan Thi Hanh Phi
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Yoseph Toni Wijaya
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Ita Novita Sari
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Kwang Seock Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Ying-Gui Yang
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Min-Woo Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Hyog Young Kwon
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
37
|
Xiao H, Xue Q, Zhang Q, Li C, Liu X, Liu J, Li H, Yang J. How Ginsenosides Trigger Apoptosis in Human Lung Adenocarcinoma Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1737-1754. [PMID: 31795742 DOI: 10.1142/s0192415x19500885] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Panax ginseng is a natural medicine that has been used globally for a long time. Moreover, several studies have reported the effective activity of ginseng in treating malignancies. Various agents containing ginseng were widely used as an antitumor treatment nowadays. Lung cancer is the most common fatal cancer in China, and lung adenocarcinoma is the most common histological type of non-small cell lung cancer (NSCLC). What's worse, many patients may have a failed response to conventional therapy including chemotherapy, radiotherapy, or molecule-targeted therapy due to drug resistance. Apoptosis is a highly ordered cellular suicidal process that plays an essential role in maintaining normal homeostasis. The pharmacological mechanism of many antineoplastic drugs involves triggering of apoptotic process. In several recent studies, ginsenosides are regarded as major active components of ginseng that have the potential to control lung cancer. Most of these results have proved that ginsenosides induce apoptosis in lung cancer cells through many different signaling pathways such as PI3K/Akt, NF-κB, EGFR, and so on. This study is aimed at reviewing the signaling pathways that underlie ginsenosides-triggered apoptotic process and encourage further studies to target promising agents against lung cancer treatment.
Collapse
Affiliation(s)
- Han Xiao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qianfei Xue
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qinghua Zhang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Chunyan Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xiaoqiu Liu
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jing Liu
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Han Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
38
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
39
|
Yang Q, Wang N, Zhang J, Chen G, Xu H, Meng Q, Du Y, Yang X, Fan H. In vitro and in silico evaluation of stereoselective effect of ginsenoside isomers on platelet P2Y 12 receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:152899. [PMID: 31454649 DOI: 10.1016/j.phymed.2019.152899] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 03/07/2019] [Accepted: 03/19/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND P2Y12 receptor (P2Y12R) is a newly discovered Gi-coupled ADP receptor that plays critical role in platelet function. Ginsenosides are the main constituents responsible for most of pharmacological actions of ginseng, especially cardio-cerebrovascular protective efficacy that is closely related to the influence on platelet function. HYPOTHESIS/PURPOSE To explore stereoselective effect of naturally abundant ginsenoside isomers, including the C-20 epimers of protopanaxadiol (PPD), protopanaxatriol (PPT), and their glycosides Rg2, Rg3, Rh1, Rh2 on P2Y12R in platelets. STUDY DESIGN/METHODS Both in vitro assay and in silico molecular docking study were performed to investigate the stereoselective effects. RESULTS In vitro assay using washed rat platelets revealed differential effects of ginsenoside isomers on ADP-induced platelet aggregation with the direction and degree of action varying with chemical structures. More to the point, the ginsenoside 20S-Rh2 but not its 20R-epimer was found to be the only one that could significantly promote in vitro platelets aggregation induced by ADP. The correlation analysis demonstrated that ginsenosides may have impact on P2Y12R related platelet functions through a cAMP-dependent pathway. Molecular docking stimulation further indicated that ginsenoside isomers could be potent substrate of P2Y12R with differential protein-ligand interaction that would be responsible for the stereoselective efficacy of C-20 ginsenoside epimers. Hydrogen bonding with Asp266 via the C-20 hydroxyl may provide ginsenosides with promoting effect on ADP-induced platelets aggregation, whereas interactions with Tyr105 could contribute to the promotion of inhibitory efficacy. CONCLUSION Ginsenosides are potent P2Y12R substrate with stereoselective effects on P2Y12R-related platelet function, which result from their chemical diversity and are closely related to the different interaction ways as P2Y12R ligand.
Collapse
Affiliation(s)
- Qianwen Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| | - Ning Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| | - Jie Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| | - Geng Chen
- School of Chemistry and Chemical Engineering, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| | - Hui Xu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China.
| | - Qingguo Meng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China.
| | - Yuan Du
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| | - Xin Yang
- School of Chemistry and Chemical Engineering, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| | - Huaying Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Yantai 264005, Shandong, China
| |
Collapse
|
40
|
Nakhjavani M, Palethorpe HM, Tomita Y, Smith E, Price TJ, Yool AJ, Pei JV, Townsend AR, Hardingham JE. Stereoselective Anti-Cancer Activities of Ginsenoside Rg3 on Triple Negative Breast Cancer Cell Models. Pharmaceuticals (Basel) 2019; 12:E117. [PMID: 31374984 PMCID: PMC6789838 DOI: 10.3390/ph12030117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
Ginsenoside Rg3 (Rg3) has two epimers, 20(S)-ginsenoside Rg3 (SRg3) and 20(R)-ginsenoside Rg3 (RRg3), and while Rg3 itself has been reported to have anti-cancer properties, few studies have been reported on the anti-cancer effects of the different epimers. The aim was to investigate the stereoselective effects of the Rg3 epimers on triple negative breast cancer (TNBC) cell lines, tested using cell-based assays for proliferation, apoptosis, cell cycle arrest, migration and invasion. Molecular docking showed that Rg3 interacted with the aquaporin 1 (AQP1) water channel (binding score -9.4 kJ mol-1). The Xenopus laevis oocyte expression system was used to study the effect of Rg3 epimers on the AQP1 water permeability. The AQP1 expression in TNBC cell lines was compared with quantitative-polymerase chain reaction (PCR). The results showed that only SRg3 inhibited the AQP1 water flux and inhibited the proliferation of MDA-MB-231 (100 μM), due to cell cycle arrest at G0/G1. SRg3 inhibited the chemoattractant-induced migration of MDA-MB-231. The AQP1 expression in MDA-MB-231 was higher than in HCC1143 or DU4475 cell lines. These results suggest a role for AQP1 in the proliferation and chemoattractant-induced migration of this cell line. Compared to SRg3, RRg3 had more potency and efficacy, inhibiting the migration and invasion of MDA-MB-231. Rg3 has stereoselective anti-cancer effects in the AQP1 high-expressing cell line MDA-MB-231.
Collapse
Affiliation(s)
- Maryam Nakhjavani
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Helen M Palethorpe
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Yoko Tomita
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Eric Smith
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Timothy J Price
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jinxin V Pei
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Amanda R Townsend
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
- Oncology Unit, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Jennifer E Hardingham
- Molecular Oncology, Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia.
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
41
|
Zhang L, Shan X, Chen Q, Xu D, Fan X, Yu M, Yan Q, Liu J. Downregulation of HDAC3 by ginsenoside Rg3 inhibits epithelial-mesenchymal transition of cutaneous squamous cell carcinoma through c-Jun acetylation. J Cell Physiol 2019; 234:22207-22219. [PMID: 31192452 DOI: 10.1002/jcp.28788] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/24/2022]
Abstract
The metastatic rate of human cutaneous squamous cell carcinoma (CSCC) has increased in recent years. Despite the current advances in therapies, effective treatments remain lacking. Ginsenoside 20(R)-Rg3 is an effective antitumor monomer extracted from ginseng, but the role of Rg3 in CSCC remains unknown. It has been reported that aberrantly elevated histone deacetylase 3 (HDAC3) is involved in tumor malignancy in multiple malignant tumors. However, the effects of HDAC3 on the regulation of c-Jun acetylation in tumor epithelial-mesenchymal transition (EMT) and migration have not been clearly illuminated. In our research, the immunohistochemistry staining results of skin tissue microarrays showed that HDAC3 staining was increased in CSCC compared with the normal dermal tissue. Then, we found that Rg3 treatment (25 and 50 μg/ml) inhibited CSCC cell (A431 and SCC12 cells) EMT through increasing E-cadherin and decreasing N-cadherin, vimentin, and Snail expression. Wound-healing and transwell assays showed that Rg3 could inhibit migration. Meanwhile, Rg3 significantly downregulated the expression of HDAC3 in CSCC cells as detected by real-time quantitative PCR, western blot, and immunofluorescence. Importantly, c-Jun acetylation was increased by the downregulation of HDAC3 with HDAC3 shRNA, and the downregulation was associated with CSCC cell EMT inhibition. Collectively, our results showed that downregulation of HDAC3 by Rg3 or shHDAC3 treatment resulted in c-Jun acetylation, which in turn inhibited CSCC cell EMT. These results indicate that HDAC3 could potentially serve as a therapeutic target therapeutic target for CSCC. Rg3 is an attractive and efficient agent that has oncotherapeutic effects and requires further investigation.
Collapse
Affiliation(s)
- Li Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xiu Shan
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qun Chen
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dayu Xu
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xinling Fan
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ming Yu
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning, China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning, China
| | - Jiwei Liu
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
42
|
Cheng Z, Xing D. Ginsenoside Rg3 inhibits growth and epithelial-mesenchymal transition of human oral squamous carcinoma cells by down-regulating miR-221. Eur J Pharmacol 2019; 853:353-363. [PMID: 30928631 DOI: 10.1016/j.ejphar.2019.03.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/21/2022]
Abstract
Ginsenoside Rg3, isolated from the roots of Panax ginseng, has been found to exert anti-cancer activity on multiple human cancers. However, there is no any literature available about the effect of Rg3 on oral squamous cell carcinoma (OSCC). This study investigated the possible anti-cancer effects of Rg3 on OSCC, as well as the possible molecular mechanisms. In vitro, cell viability and proliferation were respectively detected by CCK-8 assay and BrdU assay. Cell apoptosis was detected by Annexin V-FITC/PI assay. Cell transfection was used to change the expression of miR-221 and TIMP3. qRT-PCR and western blotting were performed to measure the expression of molecules involving in cell apoptosis, epithelial-mesenchymal transition (EMT) process, PI3K/AKT pathway and MAPK/ERK pathway. In vivo, OSCC orthotopic murine model was established and tumor volumes were measured. We found that Rg3 treatment inhibited viability, proliferation and EMT process of human OSCC SCC-9 and HSC-5 cells, but promoted cell apoptosis. miR-221 was highly expressed in OSCC tissues and cells. Rg3 reduced the expression of miR-221 in OSCC cells. Up-regulation of miR-221 abrogated the effects of Rg3 on SCC-9 and HSC-5 cell viability, proliferation, apoptosis and EMT process. TIMP3 was lowly expressed in OSCC tissues and cells, which was a direct target gene of miR-221. Rg3 inactivated PI3K/AKT and MAPK/ERK pathways in SCC-9 cells by up-regulating TIMP3. In vivo, Rg3 reduced the tumor volume of OSCC orthotopic murine model. In conclusion, Rg3 exerted anti-cancer effects on OSCC might be via down-regulating miR-221, up-regulating TIMP3, and then inactivating PI3K/AKT and MAPK/ERK pathways.
Collapse
Affiliation(s)
- Zhou Cheng
- Department of Stomatology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| | - Dayuan Xing
- Department of Stomatology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| |
Collapse
|
43
|
Ginsenoside Rg3 Inhibits Migration and Invasion of Nasopharyngeal Carcinoma Cells and Suppresses Epithelial Mesenchymal Transition. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8407683. [PMID: 30915362 PMCID: PMC6409072 DOI: 10.1155/2019/8407683] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is a highly invasive and metastatic head and neck cancer. Distant metastasis becomes the predominant mode of treatment failure in NPC patients. Ginsenoside Rg3 (Rg3), an active pharmaceutical component extracted from traditional Chinese medicine ginseng, shows antitumor effects in various cancers. In this study, we aimed to determine whether Rg3 inhibits the migration and invasion activity of NPC cells and to explore the possible mechanisms. Our results revealed that Rg3 hampers cell migration and invasion in both HNE1 and CNE2 cell lines. A reduced level of matrix metalloproteinase-2 (MMP-2) and MMP-9 was induced by Rg3 treatment. In addition, Rg3 significantly altered the expression of epithelial mesenchymal transition (EMT) markers with increased E-cadherin but decreased Vimentin and N-cadherin expression. Transforming growth factor β- (TGF-β-) induced morphological transition and marker proteins change of EMT were reversed by Rg3. What is more, Rg3 suppressed the expression of EMT-related transcription factors, especially the Zinc Finger E-Box Binding Homeobox 1 (ZEB1). In summary, our data suggested that Rg3 could inhibit migration and invasion of NPC cells. This effect of Rg3 might be mediated through regulating MMP-2 and MMP-9 expressions and suppressing EMT. Thus, Rg3 may be a potentially effective agent for the treatment of NPC.
Collapse
|
44
|
Ginsenoside Rg3: Potential Molecular Targets and Therapeutic Indication in Metastatic Breast Cancer. MEDICINES 2019; 6:medicines6010017. [PMID: 30678106 PMCID: PMC6473622 DOI: 10.3390/medicines6010017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 01/28/2023]
Abstract
Breast cancer is still one of the most prevalent cancers and a leading cause of cancer death worldwide. The key challenge with cancer treatment is the choice of the best therapeutic agents with the least possible toxicities on the patient. Recently, attention has been drawn to herbal compounds, in particular ginsenosides, extracted from the root of the Ginseng plant. In various studies, significant anti-cancer properties of ginsenosides have been reported in different cancers. The mode of action of ginsenoside Rg3 (Rg3) in in vitro and in vivo breast cancer models and its value as an anti-cancer treatment for breast cancer will be reviewed.
Collapse
|
45
|
Li J, Qi Y. Ginsenoside Rg3 inhibits cell growth, migration and invasion in Caco-2 cells by downregulation of lncRNA CCAT1. Exp Mol Pathol 2019; 106:131-138. [PMID: 30633886 DOI: 10.1016/j.yexmp.2019.01.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a troublesome disease with high morbidity and mortality. Ginsenoside Rg3 possesses anti-cancer properties. Colon Cancer Associated Transcript 1 (CCAT1) participates in the genesis, development, invasion and metastasis of colorectal cancer. In our study, we explored the effects of Rg3 on CRC cell line Caco-2 by regulating CCAT1. METHODS CRC tissue was obtained from hospital and Caco-2 cells were purchased. Caco-2 cells were treated with Rg3 and/or transfected with pc- CCAT1 or pcDNA3.1. The group without Rg3 treatment was treated as control. Cell viability, cell apoptosis, cell migration and invasion were detected by Cell Counting Kit-8 assay, flow cytometry and Transwell chamber migration/invasion assay, respectively. The expression of CyclinD1, apoptosis related proteins (p53, Bcl-2, Bax, pro-/Cleaved-Caspase-3), migration and invasion related proteins (MMP-9 and vimentin), and phosphatidylinositol 3'-kinase (PI3K)/protein kinase B (AKT) related proteins (p/t-PI3K, p/t-AKT) were examined by western blot. The expression of CCAT1 was measured by quantitative real time RCR (qRT-PCR). RESULTS Rg3 significantly decreased cell viability, migration and invasion, and promoted apoptosis. Meanwhile, the expression of Cyclin D1, matrix metalloproteinase (MMP)-9 and vimentin was downregulated. The expression of apoptosis-related proteins p53, Bax, and Cleaved-Caspase-3 were upregulated while Bcl-2 was downregulated by the treatment of Rg3 compared with control. Furthermore, CCAT1 was upregulated in CRC tissue and Rg3 negatively regulated CCAT1 expression. Transfection with pc-CCAT1 led to the opposite results as compared with transfection with pcDNA3.1 in Rg3 treated cells. In addition, Rg3 decreased the phosphorylation of PI3K and AKT. CONCLUSION Ginsenoside Rg3 inhibits migration and invasion, and promotes apoptosis of Caco-2 cells by suppression expression of LncRNA CCAT1.
Collapse
Affiliation(s)
- Jinliang Li
- Department of Anorectal Surgery, Jining No.1 People's Hospital, Jining 272011, China; Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining 272067, China
| | - Yuxi Qi
- Department of Anorectal Surgery, Jining No.1 People's Hospital, Jining 272011, China.
| |
Collapse
|
46
|
Dai Y, Wang W, Sun Q, Tuohayi J. Ginsenoside Rg3 promotes the antitumor activity of gefitinib in lung cancer cell lines. Exp Ther Med 2018; 17:953-959. [PMID: 30651886 DOI: 10.3892/etm.2018.7001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/05/2018] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is one of the most common types of cancer with one of the highest incidence and mortality rates. Gefitinib is widely used for the treatment of non-small cell lung cancer (NSCLC). However, issues regarding drug resistance, toxicity and limited applicability have been associated with gefitinib. The aim of the present study was to investigate whether ginsenoside Rg3 enhances the anticancer activity of gefitinib in NSCLC cells. MTT assay demonstrated that ginsenoside Rg3 increased the cytotoxic effect of gefitinib in NSCLC cell lines in a dose- and time-dependent manner. In addition, flow cytometric analysis revealed that the combined treatment with gefitinib and ginsenoside Rg3 significantly increased apoptosis in NSCLC cell lines. Transwell migration assays demonstrated that the combined treatment with gefitinib and ginsenoside Rg3 significantly decreased NSCLC cell migration compared with gefitinib or ginsenoside Rg3 treatment alone. Furthermore, western blot analysis revealed that in NSCLC cell lines, the combined treatment with gefitinib and ginsenoside Rg3 increased protein expression levels of pro-apoptotic proteins Bax and cleaved-caspase-3, whilst the expression level of anti-apoptotic protein Bcl-2 decreased. In addition, western blot analysis revealed that, in NSCLC cell lines, the combined treatment with gefitinib and ginsenoside Rg3 decreased the protein expression levels of pro-migration factors SNAIL and SLUG, whilst the expression level of anti-migration protein E-cadherin increased. In conclusion, ginsenoside Rg3 may be able to enhance the anticancer activity of gefitinib, making NSCLC cells more sensitive to gefitinib.
Collapse
Affiliation(s)
- Yuemei Dai
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Wenran Wang
- Department of Cancer, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Qingchao Sun
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Jiazina Tuohayi
- Department of Respiratory Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
47
|
Sun MY, Song YN, Zhang M, Zhang CY, Zhang LJ, Zhang H. Ginsenoside Rg3 inhibits the migration and invasion of liver cancer cells by increasing the protein expression of ARHGAP9. Oncol Lett 2018; 17:965-973. [PMID: 30655855 PMCID: PMC6313058 DOI: 10.3892/ol.2018.9701] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/10/2018] [Indexed: 12/13/2022] Open
Abstract
Ginsenoside Rg3, a naturally occurring phytochemical, serves an important role in the prevention and treatment of cancer. In the present study, with the aim to reveal the molecular mechanism of Rg3 in liver cancer cell metastasis, the anti-migration and anti-invasion effects of Rg3 on liver cancer cells were investigated. It was demonstrated that Rg3 caused marked inhibition of cell migration and invasion of human liver cancer cells, HepG2 and MHCC-97L, in vitro, and the growth of HepG2 and MHCC-97L tumors in BABL/c nude mice. The protein expression of Rho GTPase activating protein 9 (ARHGAP9) was increased both in HepG2 and MHCC-97L cells. Following ARHGAP9 knockdown, the results of Transwell and tumorigenesis assays revealed that the anti-migration, anti-invasion and anti-tumor growth effects of Rg3 were impaired significantly. The increased expression of ARHGAP9 protein induced by Rg3 was remarkably suppressed. All results suggested that ARHGAP9 protein may be a vital regulator in the anti-metastatic role of Rg3. To the best of our knowledge, the present study is the first to report that Rg3 effectively suppressed the migration and invasion of liver cancer cells by upregulating the protein expression of ARHGAP9, indicating a novel natural therapeutic agent and a therapeutic target for the treatment of liver cancer.
Collapse
Affiliation(s)
- Meng-Yao Sun
- Department of Pharmaceutical Botany, School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Ya-Nan Song
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Miao Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Chun-Yan Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Li-Jun Zhang
- Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| | - Hong Zhang
- Department of Pharmaceutical Botany, School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China.,Central Laboratory, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, P.R. China
| |
Collapse
|
48
|
Yang J, Li S, Wang L, Du F, Zhou X, Song Q, Zhao J, Fang R. Ginsenoside Rg3 Attenuates Lipopolysaccharide-Induced Acute Lung Injury via MerTK-Dependent Activation of the PI3K/AKT/mTOR Pathway. Front Pharmacol 2018; 9:850. [PMID: 30116194 PMCID: PMC6082957 DOI: 10.3389/fphar.2018.00850] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical disease with high morbidity in both humans and animals. Ginsenoside Rg3, a type of traditional Chinese medicine extracted from ginseng, is widely used to cure many inflammation-related diseases. However, the specific molecular mechanism of the effects of ginsenoside Rg3 on inflammation has rarely been reported. Thus, we established a mouse model of lipopolysaccharide (LPS)-induced ALI to investigate the immune protective effects of ginsenoside Rg3 and explore its molecular mechanism. In wild type (WT) mice, we found that ginsenoside Rg3 treatment significantly mitigated pathological damages and reduced myeloperoxidase (MPO) activity as well as the production of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6); furthermore, the production of anti-inflammatory mediators interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), polarization of M2 macrophages and expression levels of the phosphorylation of phosphatidylinositol 3-hydroxy kinase (PI3K), protein kinase B (PKB, also known as AKT), mammalian target of rapamycin (mTOR) and Mer receptor tyrosine kinase (MerTK) were promoted. However, there were no significant differences with regards to the pathological damage, MPO levels, inflammatory cytokine levels, and protein expression levels of the phosphorylation of PI3K, AKT and mTOR between the LPS treatment group and ginsenoside Rg3 group in MerTK-/- mice. Taken together, the present study demonstrated that ginsenoside Rg3 could attenuate LPS-induced ALI by decreasing the levels of pro-inflammatory mediators and increasing the production of anti-inflammatory cytokines. These processes were mediated through MerTK-dependent activation of its downstream the PI3K/AKT/mTOR pathway. These findings identified a new site of the specific anti-inflammatory mechanism of ginsenoside Rg3.
Collapse
Affiliation(s)
- Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Senyang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Luyao Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fen Du
- Hubei Center for Animal Diseases Control and Prevention, Wuhan, China
| | - Xiaoliu Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiqi Song
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
49
|
Kee JY, Han YH, Mun JG, Um JY, Hong SH. Pharmacological effect of prohibited combination pair Panax ginseng and Veratrum nigrum on colorectal metastasis in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2018; 220:177-187. [PMID: 29601980 DOI: 10.1016/j.jep.2018.03.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/21/2018] [Accepted: 03/26/2018] [Indexed: 05/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The pharmacological effect derived from herb-herb interaction is important to constitute the prescription especially in traditional oriental medicine. The relationship of two medicinal herbs is called "couplet medicinals" which is used in pair for the purpose of enhancing the therapeutic effect, reducing the toxic effect or the adverse effect. The "Eighteen Incompatible Medicaments" constitute one of the contents in the incompatibility of traditional oriental drugs in a prescription. Among the "Eighteen Incompatible Medicaments", the roots and rhizomes of Veratrum nigrum (VN), is incompatible with the roots and rhizomes of Panax ginseng (PG). However, definite evidences of adverse effect by these combinations has yet to be reported. MATERIALS AND METHODS The aim of the present study was to investigate the effects of ethanol extracts of PG, VN, and their combination (P + V) on the metastatic ability of colorectal cancer (CRC) cells using WST assay, flow cytometry, western blot analysis, real-time RT-PCR, immunofluorescence, migration assay, invasion assay, zymography, and an in vivo experiment with a lung-metastasis mouse model. RESULTS The PG extract decreased cell proliferation by inducing cell cycle arrest and apoptosis of CRC cells. In addition, PG inhibited metastatic abilities of CRC cells including Epithelial-Mesenchymal Transition, migration, and invasion. Additionally, the PG extract suppressed lung metastasis of the CRC cells in the mouse model. However, the P + V extract exhibited weaker anti-proliferative and anti-metastatic effects than PG alone. CONCLUSION Based on these results, the P + V couplet medicinal attenuates the anti-metastatic effects of PG, both in vitro and in vivo.
Collapse
Affiliation(s)
- Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yo-Han Han
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jeong-Geon Mun
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jae-Young Um
- Department of Pharmacology, College of Korean Medicine, Institute of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea.
| |
Collapse
|
50
|
Sun B, Hu C, Yang Z, Zhang X, Zhao L, Xiong J, Ma J, Chen L, Qian H, Luo X, Shi L, Li J, Cheng X, Yin Z. Midkine promotes hepatocellular carcinoma metastasis by elevating anoikis resistance of circulating tumor cells. Oncotarget 2018; 8:32523-32535. [PMID: 28430645 PMCID: PMC5464806 DOI: 10.18632/oncotarget.15808] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/15/2017] [Indexed: 02/07/2023] Open
Abstract
Midkine is overexpressed in hepatocellular carcinoma (HCC) and plays a role in tumor progression, but less is known about its role in resistance of circulating tumor cells (CTCs) to anoikis which leading to recurrence and metastasis. The aim of the present study was to analyze whether midkine was associated with HCC progression with anoikis resistance. We found that cultured HCC cells were more resistant to anoikis, which paralleled midkine expression, and midkine treatment significantly inhibited anoikis in a dose-dependent manner. Furthermore, in in vitro and in vivo assays, knockdown of midkine resulted in significant sensitivity to anoikis, decreased cell survival and significantly decreased tumor occurrence rate. Patients with midkine-elevated HCC had higher CTC counts and less apoptotic CTCs, as well as significantly higher recurrence rate and shorter recurrence-free interval. To understand the molecular mechanism underlying the midkine with HCC progression, we performed in vitro and in vivo studies. We found that midkine plays an important role in enhancement of HCC cell resistance to anoikis, thereby promoting subsequent metastasis. Activation of PI3K/Akt/NF-κB/TrkB signaling by midkine-activated anaplastic lymphomakinase (ALK) is responsible for anoikis resistance.
Collapse
Affiliation(s)
- Bin Sun
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Congli Hu
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Zhibin Yang
- Colorectal Cancer Clinical Research Center, Third Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaofeng Zhang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Linlin Zhao
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Junye Xiong
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Junyong Ma
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lei Chen
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Haihua Qian
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xiangji Luo
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lehua Shi
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jun Li
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xianshuo Cheng
- Colorectal Cancer Clinical Research Center, Third Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Zhengfeng Yin
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|