1
|
Li D, Zhong Z, Ko CN, Tian T, Yang C. From mundane to classic: Sinomenine as a multi-therapeutic agent. Br J Pharmacol 2025; 182:2159-2180. [PMID: 37846470 DOI: 10.1111/bph.16267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/10/2023] [Accepted: 10/08/2023] [Indexed: 10/18/2023] Open
Abstract
Sinomenine is an active substance extracted from the traditional Chinese medicine Sinomenium acutum. Sinomenine has been shown to mediate a wide range of pharmacological actions and is known to possess good anti-inflammatory, immunosuppressive, antitumor, neuroprotective, antiarrhythmic and other pharmacological effects. Understanding the underlying mechanisms and the association between the targets and the pharmaceutical effects on different diseases is crucial to the discovery and design of new treatment strategies. In this review, we aim to give a systematic and comprehensive overview of the research progress of sinomenine over the past 20 years. We first describe the metabolism of sinomenine in vivo and then summarize the pharmacological actions of sinomenine on different diseases. Furthermore, the potential binding properties of sinomenine and the potential of developing new sinomenine-based drugs are also reviewed. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Chung-Nga Ko
- The International Eye Research Institute of The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Tiantian Tian
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, China
| | - Chao Yang
- National Engineering Research Center For Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| |
Collapse
|
2
|
Bi L, Wang X, Li J, Li W, Wang Z. Epigenetic modifications in early stage lung cancer: pathogenesis, biomarkers, and early diagnosis. MedComm (Beijing) 2025; 6:e70080. [PMID: 39991629 PMCID: PMC11843169 DOI: 10.1002/mco2.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
The integration of liquid biopsy with epigenetic markers offers significant potential for early lung cancer detection and personalized treatment. Epigenetic alterations, including DNA methylation, histone modifications, and noncoding RNA changes, often precede genetic mutations and are critical in cancer progression. In this study, we explore how liquid biopsy, combined with epigenetic markers, can provide early detection of lung cancer, potentially predicting onset up to 4 years before clinical diagnosis. We discuss the challenges of targeting epigenetic regulators, which could disrupt cellular balance if overexploited, and the need for maintaining key gene expressions in therapeutic applications. This review highlights the promise and challenges of using liquid biopsy and epigenetic markers for early-stage lung cancer diagnosis, with a focus on optimizing treatment strategies for personalized and precision medicine.
Collapse
Affiliation(s)
- Lingfeng Bi
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xin Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jiayi Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan UniversityChengduSichuanChina
- The Research Units of West China, Chinese Academy of Medical SciencesWest China HospitalChengduSichuanChina
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease‐related Molecular Network, State Key Laboratory of Respiratory Health and MultimorbidityWest China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Respiratory Health, Frontiers Science Center for Disease‐Related Molecular NetworkWest China Hospital, Sichuan UniversityChengduSichuanChina
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan UniversityChengduSichuanChina
- The Research Units of West China, Chinese Academy of Medical SciencesWest China HospitalChengduSichuanChina
| |
Collapse
|
3
|
Xi Z, Dai R, Ze Y, Jiang X, Liu M, Xu H. Traditional Chinese medicine in lung cancer treatment. Mol Cancer 2025; 24:57. [PMID: 40001110 PMCID: PMC11863959 DOI: 10.1186/s12943-025-02245-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Lung cancer remains a major global health challenge and one of the leading causes of cancer-related deaths worldwide. Despite significant advancements in treatment, challenges such as drug resistance, side effects, metastasis and recurrence continue to impact patient outcomes and quality of life. In response, there is growing interest in complementary and integrative approaches to cancer care. Traditional Chinese medicine (TCM), with its long history, abundant clinical experience, holistic perspective and individualized approach, has garnered increasing attention for its role in lung cancer prevention and management. This review provides a comprehensive overview of the advances in TCM for lung cancer treatment, covering its theoretical foundation, treatment principles, clinical experiences and evidence supporting its efficacy. We also provide a systematic summary of the preclinical mechanisms, through which TCM impacts lung cancer, including the induction of cell death, reversal of drug resistance, inhibition of metastasis and modulation of immune responses. Additionally, future prospects for TCM in lung cancer treatment are discussed, offering insights into its expanded application and integration with modern medicine to address this challenging disease.
Collapse
Affiliation(s)
- Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Yufei Ze
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Xue Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China
| | - Mengfan Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China.
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, China.
| |
Collapse
|
4
|
Ling F, Xie W, Kui X, Cai Y, He M, Ma J. miR-141-3p inhibited BPA-induced proliferation and migration of lung cancer cells through PTGER4. Cytotechnology 2025; 77:28. [PMID: 39741890 PMCID: PMC11683044 DOI: 10.1007/s10616-024-00692-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
The chemical substance bisphenol A (BPA) is widely used in household products, and its effect on human health has frequently been the focus of research. The aim of this study was to explore the potential molecular regulatory mechanism of BPA on the proliferation and migration of lung cancer cells. In this study, the H1299 and A549 lung cancer cell lines were selected as the study objects. The cells were treated with different concentrations of BPA (0, 0.1, 1, or 10 μM), and cell viability, proliferation, and migration were evaluated by CCK-8, EdU, clonogenic, and scratch test assays. Western blotting and RT‒qPCR were used to detect the expression of related proteins and genes. Our findings indicated that BPA markedly enhanced both the proliferation and migration capacities of lung cancer cells. In BPA-treated lung cancer cells, the level of miR-141-3p was decreased, PTGER4 expression was significantly increased, and PTGER4 knockdown reduced BPA-induced lung cancer cell proliferation and migration. In addition, miR-141-3p can target and negatively regulate the expression of PTGER4 and further inhibit PI3K/AKT signaling pathway activation and MMPs expression. Moreover, PTGER4 overexpression weakened the inhibitory effect of the miR-141-3p mimic on the proliferation and migration of lung cancer cells. In conclusion, miR-141-3p can inhibit the proliferation and migration of BPA-induced lung cancer cells by downregulating PTGER4, providing a new potential target for the treatment and prevention of lung cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00692-5.
Collapse
Affiliation(s)
- Feng Ling
- Thoracic Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101 Yunnan China
| | - Wenbo Xie
- Digestive System Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101 Yunnan China
| | - Xiang Kui
- Pathology Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101 Yunnan China
| | - Yuyin Cai
- Thoracic Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101 Yunnan China
| | - Meng He
- Thoracic Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101 Yunnan China
| | - Jianqiang Ma
- Thoracic Surgery Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101 Yunnan China
| |
Collapse
|
5
|
Yu F, Peng Z, Gao N, Tang Z, Liao Z, Zhao S, Zhong S, Umwiza G, Huang H, Long W, He Z. Sinomenine attenuates uremia vascular calcification by miR-143-5p. Sci Rep 2025; 15:1798. [PMID: 39806038 PMCID: PMC11730593 DOI: 10.1038/s41598-025-86055-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Vascular calcification is considered to be a killer of the cardiovascular system, involved inflammation and immunity. There is no approved therapeutic strategy for the prevention of vascular calcification. Sinomenine exhibited anti-inflammatory and immunosuppressive effects. Objective of this study was to investigate the effect of sinomenine in vascular calcification and its potential molecular mechanism. Adenine-induced uremic rats were constructed and administrated with sinomenine. Optical clearing of aortas, alizarin red staining, von Kossa staining, calcification quantification, micro-CT analyses of vascular calcification were performed to analyze calcification in aortas. Administration of 40 mg/kg/d sinomenine effectively alleviated vascular calcification in uremic rats. The miRNA sequencing revealed differentially expressed miRNAs in aortas and bioinformatic analysis assisted with miRNA screening. We screened 9 differential expressed miRNAs and their predicted target genes. By qRT-PCR, we validated that the expression of rno-miR-143-5p was corresponding to our prediction. Sinomenine inhibited vascular smooth muscle cells (VSMCs) calcification, accompanied with miR-143-5p upregulation. MiR-143-5p mimic decreased VSMCs calcification in high phosphate condition. On the contrary, miR-143-5p inhibitor increased VSMCs calcification in high phosphate condition, which was inhibited by sinomenine. In chronic kidney disease patients with vascular calcification, the expression level of circulating miR-143-5p was lower than those without vascular calcification. Sinomenine significantly inhibited vascular calcification in VSMCs and uremic rat. MiR-143-5p was one of the collection of miRNAs modified by sinomenine in vascular calcification. Reduction of miR-143-5p in VSMCs was not only a concomitant phenomenon in pro-calcification condition but also contribute to VSMCs calcification. Circulating miR-143-5p was supposed to be a potential biomarker for vascular calcification in chronic kidney disease patients. In conclusion, sinomenine effectively alleviated vascular calcification, which was attributed to miR-143-5p regulation partly.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Morphinans/pharmacology
- Animals
- Vascular Calcification/drug therapy
- Vascular Calcification/genetics
- Vascular Calcification/etiology
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Uremia/complications
- Uremia/drug therapy
- Uremia/genetics
- Rats
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Humans
- Rats, Sprague-Dawley
- Aorta/pathology
- Aorta/metabolism
- Aorta/drug effects
- Disease Models, Animal
Collapse
Affiliation(s)
- Fengyi Yu
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Department of Gastroenterology, Yiyang Central Hospital, Yiyang, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhong Peng
- Department of Gastroenterology, Yiyang Central Hospital, Yiyang, Hunan, China
| | - Ning Gao
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
| | - Zixu Tang
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zihao Liao
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Song Zhao
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shuzhu Zhong
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Gloria Umwiza
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Huang
- Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Long
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China
| | - Zhangxiu He
- Department of Nephrology, Yiyang Central Hospital, 118 Kangfubei Road, Yiyang, 413000, Hunan, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
6
|
Silvia Lima RQD, Vasconcelos CFM, Gomes JPA, Bezerra de Menezes EDS, de Oliveira Silva B, Montenegro C, Paiva Júnior SDSL, Pereira MC. miRNA-21, an oncomiR that regulates cell proliferation, migration, invasion and therapy response in lung cancer. Pathol Res Pract 2024; 263:155601. [PMID: 39413459 DOI: 10.1016/j.prp.2024.155601] [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: 03/18/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024]
Abstract
Lung cancer is the leading cause of cancer-related death globally, with poor survival rates due mostly to a lack of early detection. The usual diagnostic technique includes a biopsy, which is frequently performed later in the disease's progression. In order to uncover processes that improve illness detection and prognosis, miRNA-21 emerges as a major miRNA identified in a variety of cancer types, including lung cancer. This review compiles insights into the involvement of miRNA-21 within the distinct cellular processes underlying lung cancer. To achieve this, we conducted an extensive literature review, drawing from published in vitro, in vivo and clinical trials studies. Searches were performed in the PubMed, Scielo, CAPES Journal Portal, BVS, INCA, and Clinical Trials.Gov. Only English written articles were selected. As screening criteria, we selected articles that explored the modulation pathways of miRNA-21, along with the proteins and genes implicated in tumorigenesis, metastasis, therapy resistance to established treatments, and their significance in the diagnosis and prognosis of lung cancer. A total of 3294 articles were identified, and 37 papers were selected to compose the review, after analysing selection criteria. Of these, 57 % studies presented in vitro evaluation, 22 % studies showed in vivo analysis, and 12 clinical trials were found. This study elucidates the principal signaling pathways influenced by miRNA-21, which play a pivotal role in lung cancer development. This comprehensive review sheds light on the potential significance of miRNA-21 as a critical mechanism for improving the prognosis of lung cancer patients, facilitating the transition of experimental data into the clinical phase. Therefore, we summarized published articles of miRNA-21 modulated signal pathways in lung cancer.
Collapse
Affiliation(s)
| | | | - João Pedro Alves Gomes
- Research Center for Therapeutic Innovation Suely Galdino, Federal University of Pernambuco, Recife, Brazil
| | | | - Barbara de Oliveira Silva
- Research Center for Therapeutic Innovation Suely Galdino, Federal University of Pernambuco, Recife, Brazil
| | - Claudio Montenegro
- Research Center for Therapeutic Innovation Suely Galdino, Federal University of Pernambuco, Recife, Brazil
| | | | - Michelly Cristiny Pereira
- Research Center for Therapeutic Innovation Suely Galdino, Federal University of Pernambuco, Recife, Brazil.
| |
Collapse
|
7
|
Chen J, Wang B, Meng T, Li C, Liu C, Liu Q, Wang J, Liu Z, Zhou Y. Oxidative Stress and Inflammation in Myocardial Ischemia-Reperfusion Injury: Protective Effects of Plant-Derived Natural Active Compounds. J Appl Toxicol 2024. [PMID: 39482870 DOI: 10.1002/jat.4719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of death among patients with cardiovascular diseases. Percutaneous coronary intervention (PCI) has been the preferred clinical treatment for AMI due to its safety and efficiency. However, research indicates that the rapid restoration of myocardial oxygen supply following PCI can lead to secondary myocardial injury, termed myocardial ischemia-reperfusion injury (MIRI), posing a grave threat to patient survival. Despite ongoing efforts, the mechanisms underlying MIRI are not yet fully elucidated. Among them, oxidative stress and inflammation stand out as critical pathophysiological mechanisms, playing significant roles in MIRI. Natural compounds have shown strong clinical therapeutic potential due to their high efficacy, availability, and low side effects. Many current studies indicate that natural compounds can mitigate MIRI by reducing oxidative stress and inflammatory responses. Therefore, this paper reviews the mechanisms of oxidative stress and inflammation during MIRI and the role of natural compounds in intervening in these processes, aiming to provide a basis and reference for future research and development of drugs for treating MIRI.
Collapse
Affiliation(s)
- Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Zhang Y, Ran H, Hui S, Qian L. Effects of sinomenine on a rat orthotopic liver carcinoma model. Histol Histopathol 2024; 39:795-804. [PMID: 38084512 DOI: 10.14670/hh-18-683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
OBJECTIVE Liver carcinoma is a common malignant tumor. In this study, an orthotopic liver carcinoma model was established by B-ultrasound, and the therapeutic effect of sinomenine (Sin) on the disease was investigated. METHODS SD rats were randomly divided into control, Sin, Sorafenib (Sor), and combination (Sin+Sor) groups (n=8). An orthotopic liver carcinoma model was established by inoculating N1-S1 cells into the rat liver by B-ultrasound-guided, and tumor volume was monitored three times by B-ultrasound after inoculation. After drug treatment, the tumor tissues were stained with HE and TUNEL, and the levels of inflammatory cytokines, ALT and AST were detected by ELISA. The numbers of erythrocytes, leukocytes and platelets were detected. Immunohistochemistry and immunofluorescence were used to detect the expression of Ki-67, CD44, VEGF and CD31. The levels of cell cycle, apoptosis-related proteins were detected by western blot. RESULTS B-ultrasound monitoring found that Sin reduced tumor volume. Moreover, Sin improved tissue lesions, and promoted cancer cell apoptosis. Sin decreased the levels of inflammatory cytokines, AST and ALT, and decreased the numbers of erythrocytes, leukocytes and platelets. Simultaneously, the expressions of Ki-67, CD44, VEGF and CD31 were decreased in the Sin group. Furthermore, Sin decreased the Bcl-2, Cyclin D1, CDK4, CDK6 and Survivin levels, but increased Bax, Cleaved-caspase3/pro-caspase3, P21 and P27 levels. More importantly, the combination of Sin and Sor treatment was more effective than treatment alone. CONCLUSION A rat orthotopic liver carcinoma model was established under the guidance of B-ultrasound, and Sin had a therapeutic effect on orthotopic liver carcinoma.
Collapse
Affiliation(s)
- Yunlong Zhang
- Department of Ultrasonography, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China
| | - Hongmei Ran
- Department of Ultrasonography, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China
| | - Shanshan Hui
- Department of Ultrasonography, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China
| | - Liping Qian
- Department of Radiology, Hangzhou Cancer Hospital, Hangzhou, PR China.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Arunrungvichian K, Vajragupta O, Hayakawa Y, Pongrakhananon V. Targeting Alpha7 Nicotinic Acetylcholine Receptors in Lung Cancer: Insights, Challenges, and Therapeutic Strategies. ACS Pharmacol Transl Sci 2024; 7:28-41. [PMID: 38230275 PMCID: PMC10789132 DOI: 10.1021/acsptsci.3c00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/26/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024]
Abstract
Alpha7 nicotinic acetylcholine receptor (α7 nAChR) is an ion-gated calcium channel that plays a significant role in various aspects of cancer pathogenesis, particularly in lung cancer. Preclinical studies have elucidated the molecular mechanism underlying α7 nAChR-associated lung cancer proliferation, chemotherapy resistance, and metastasis. Understanding and targeting this mechanism are crucial for developing therapeutic interventions aimed at disrupting α7 nAChR-mediated cancer progression and improving treatment outcomes. Drug research and discovery have determined natural compounds and synthesized chemical antagonists that specifically target α7 nAChR. However, approved α7 nAChR antagonists for clinical use are lacking, primarily due to challenges related to achieving the desired selectivity, efficacy, and safety profiles required for effective therapeutic intervention. This comprehensive review provided insights into the molecular mechanisms associated with α7 nAChR and its role in cancer progression, particularly in lung cancer. Furthermore, it presents an update on recent evidence about α7 nAChR antagonists and addresses the challenges encountered in drug research and discovery in this field.
Collapse
Affiliation(s)
- Kuntarat Arunrungvichian
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Unit
of Compounds Library for Drug Discovery, Mahidol University, Bangkok 10400, Thailand
| | - Opa Vajragupta
- Research
Affairs, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Yoshihiro Hayakawa
- Institute
of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Varisa Pongrakhananon
- Department
of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Preclinical
Toxicity and Efficacy Assessment of Medicines and Chemicals Research
Unit, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
12
|
He Z, Wang Y, Han L, Hu Y, Cong X. The mechanism and application of traditional Chinese medicine extracts in the treatment of lung cancer and other lung-related diseases. Front Pharmacol 2023; 14:1330518. [PMID: 38125887 PMCID: PMC10731464 DOI: 10.3389/fphar.2023.1330518] [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: 10/31/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Lung cancer stands as one of the most prevalent malignancies worldwide, bearing the highest morbidity and mortality rates among all malignant tumors. The treatment of lung cancer primarily encompasses surgical procedures, radiotherapy, and chemotherapy, which are fraught with significant side effects, unfavorable prognoses, and a heightened risk of metastasis and relapse. Although targeted therapy and immunotherapy have gradually gained prominence in lung cancer treatment, diversifying the array of available methods, the overall recovery and survival rates for lung cancer patients remain suboptimal. Presently, with a holistic approach and a focus on syndrome differentiation and treatment, Traditional Chinese Medicine (TCM) has emerged as a pivotal player in the prognosis of cancer patients. TCM possesses characteristics such as targeting multiple aspects, addressing a wide range of concerns, and minimizing toxic side effects. Research demonstrates that Traditional Chinese Medicine can significantly contribute to the treatment or serve as an adjunct to chemotherapy for lung cancer and other lung-related diseases. This is achieved through mechanisms like inhibiting tumor cell proliferation, inducing tumor cell apoptosis, suppressing tumor angiogenesis, influencing the cellular microenvironment, regulating immune system function, impacting signal transduction pathways, and reversing multidrug resistance in tumor cells. In this article, we offer an overview of the advancements in research concerning Traditional Chinese Medicine extracts for the treatment or adjunctive chemotherapy of lung cancer and other lung-related conditions. Furthermore, we delve into the challenges that Traditional Chinese Medicine extracts face in lung cancer treatment, laying the foundation for the development of diagnostic, prognostic, and therapeutic targets.
Collapse
Affiliation(s)
- Zhenglin He
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Yihan Wang
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| | - Liang Han
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yue Hu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianling Cong
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China
- Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Yan J, Yang J, Shen H, Gao R, Lv S. Sinomenine regulates circTRPM7-related pathway to inhibit gastric cancer cell growth and metastasis. Chem Biol Drug Des 2023; 102:870-881. [PMID: 37495546 DOI: 10.1111/cbdd.14297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/24/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Sinomenine has been found to have antitumor effects in a variety of cancers, including gastric cancer. Circular RNA (circRNA) is an important regulator of gastric cancer progression. However, it is not known whether Sinomenine mediates gastric cancer processes by regulating circRNA-related pathways. Quantitative real-time PCR was used to measure the expression of circTRPM7, microRNA-145-5p (miR-145-5p), and pre-B-cell leukemia homeobox 3 (PBX3). MTT assay, colony formation assay, EdU assay, transwell assay, wound-healing assay, and flow cytometry were used to detect cell proliferation, migration, invasion, and apoptosis. The expression of related proteins was detected by Western blot. Mechanically, the interaction of miR-145-5p with circTRPM7/PBX3 was validated by dual-luciferase reporter assay and RIP assay. Our study showed that circTRPM7 expression was reduced in Sinomenine-treated gastric cancer cells. Moreover, overexpression of circTRPM7 upregulated the growth and metastasis of Sinomenine-treated gastric cancer cells. CircTRPM7 could sponge miR-145-5p, and miR-145-5p reversed the effect of circTRPM7 on the growth and metastasis of Sinomenine-treated gastric cancer cells. PBX3 was the target of miR-145-5p, and knockdown of PBX3 could restore the in-miR-145-5p promotion effect on the malignant behavior of Sinomenine-treated gastric cancer cells. To sum up, our data indicated that Sinomenine played an antitumor role in gastric cancer cells via circTRPM7/miR-145-5p/PBX3 axis.
Collapse
Affiliation(s)
- Jingwei Yan
- Department of Traditional Chinese Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Jingqing Yang
- Department of Geriatric Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Huifen Shen
- Department of Traditional Chinese Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Rong Gao
- Department of Rheumatology and Immunology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Shilong Lv
- Traditional Chinese Medicine, Weihai Health School, Weihai, China
| |
Collapse
|
14
|
Zhu J, Zhu H, Gao J. The anti-tumor potential of sinomenine: a narrative review. Transl Cancer Res 2023; 12:2393-2404. [PMID: 37859743 PMCID: PMC10583013 DOI: 10.21037/tcr-23-267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/14/2023] [Indexed: 10/21/2023]
Abstract
Background and Objective Currently, chemotherapy is the main treatment for most tumors. However, drug resistance and many adverse reactions associated with chemotherapy greatly limit its use. Therefore, an increasing number of researchers have shifted the research focus the anti-tumor activity of traditional Chinese medicine. The objective of this article is to review the anti-tumor mechanism of sinomenine and its derivatives to provide a reference for further study and clinical transformation. Methods In this study, we searched for relevant articles on the anti-tumor mechanism of Sinomenium using databases such as PubMed and Medline. Key Content and Findings Sinomenine is a monomer alkaloid component extracted from the rhizome of Sinomenium acuturn. A number of basic studies have proven that sinomenine and its derivatives show significant anti-tumor activity in breast cancer, lung cancer, liver cancer, stomach cancer, ovarian cancer, osteosarcoma and other tumors. They can induce apoptosis and autophagic death of tumor cells, inhibit proliferation, migration and invasion of tumor cells, increase the sensitivity of tumor cells to radiotherapy and chemotherapy, and reverse the drug resistance through various molecular mechanisms. In addition, sinomenine can effectively relieve osteolysis and bone pain in tumor patients. At present, anti-tumor research on sinomenine remains in the basic experimental stage. Conclusions Sinomenine and its derivatives are rich in substances with high anti-tumor potential. This analysis provides a review of the anti-tumor effects and mechanisms of sinomenine, with the hope of further exploring the medical value of sinomenine in anti-tumor treatments.
Collapse
Affiliation(s)
- Jun Zhu
- The Third Affiliated Hospital of Nanchang University, The First Hospital of Nanchang City, Nanchang, China
| | - Hong Zhu
- Department of Gynecology, Jiangxi Cancer Hospital, Nanchang, China
| | - Jun Gao
- Department of Gynecology, Jiangxi Cancer Hospital, Nanchang, China
| |
Collapse
|
15
|
Bayat H, Pourgholami MH, Rahmani S, Pournajaf S, Mowla SJ. Synthetic miR-21 decoy circularized by tRNA splicing mechanism inhibited tumorigenesis in glioblastoma in vitro and in vivo models. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:432-444. [PMID: 37181451 PMCID: PMC10173299 DOI: 10.1016/j.omtn.2023.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023]
Abstract
Glioblastoma multiforme (GBM) is the deadliest primary central nervous system tumor. miRNAs (miRs), a class of non-coding RNAs, are considered pivotal post-transcriptional regulators of cell signaling pathways. miR-21 is a reliable oncogene that promotes tumorigenesis of cancer cells. We first performed an in silico analysis on 10 microarray datasets retrieved from TCGA and GEO databases to elucidate top differentially expressed miRs. Furthermore, we generated a circular miR-21 decoy, CM21D, using the tRNA-splicing mechanism in GBM cell models, U87 and C6. The inhibitory efficacy of CM21D with that of a linear form, LM21D, was compared under in vitro conditions and an intracranial C6 rat glioblastoma model. miR-21 significantly overexpressed in GBM samples and confirmed in GBM cell models using qRT-PCR. CM21D was more efficient than LM21D at inducing apoptosis, inhibiting cell proliferation and migration, and interrupting the cell cycle by restoring the expression of miR-21 target genes at RNA and protein levels. Moreover, CM21D suppressed tumor growth more effectively than LM21D in the C6-rat GBM model (p < 0.001). Our findings validate miR-21 as a promising therapeutic target for GBM. The introduced CM21D by sponging miR-21 reduced tumorigenesis of GBM and can be considered a potential RNA-base therapy to inhibit cancers.
Collapse
Affiliation(s)
- Hadi Bayat
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-111, Iran
| | | | - Saeid Rahmani
- School of Computer Science, Institute for Research in Fundamental Sciences (IPM), Tehran 19538-33511, Iran
| | - Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-111, Iran
| |
Collapse
|
16
|
Almuntashiri S, Alhumaid A, Zhu Y, Han Y, Dutta S, Khilji O, Zhang D, Wang X. TIMP-1 and its potential diagnostic and prognostic value in pulmonary diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:67-76. [PMID: 38343891 PMCID: PMC10857872 DOI: 10.1016/j.pccm.2023.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Indexed: 08/02/2024]
Abstract
Tissue inhibitors of metalloproteases (TIMPs) have caught the attention of many scientists due to their role in various physiological and pathological processes. TIMP-1, 2, 3, and 4 are known members of the TIMPs family. TIMPs exert their biological effects by, but are not limited to, inhibiting the activity of metalloproteases (MMPs). The balance between MMPs and TIMPs is critical for maintaining homeostasis of the extracellular matrix (ECM), while the imbalance between MMPs and TIMPs can lead to pathological changes, such as cancer. In this review, we summarized the current knowledge of TIMP-1 in several pulmonary diseases namely, acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), pneumonia, asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis, and pulmonary fibrosis. Considering the potential of TIMP-1 serving as a non-invasive diagnostic and/or prognostic biomarker, we also reviewed the circulating TIMP-1 levels in translational and clinical studies.
Collapse
Affiliation(s)
- Sultan Almuntashiri
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 55473, Saudi Arabia
| | - Abdullah Alhumaid
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 55473, Saudi Arabia
| | - Yin Zhu
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Yohan Han
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Ohmed Khilji
- Department of Emergency Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| |
Collapse
|
17
|
Wang S, Zhang L, Zhou Y, Huang J, Zhou Z, Liu Z. A review on pharmacokinetics of sinomenine and its anti-inflammatory and immunomodulatory effects. Int Immunopharmacol 2023; 119:110227. [PMID: 37119677 DOI: 10.1016/j.intimp.2023.110227] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Autoimmune diseases (ADs), with significant effects on morbidity and mortality, are a broad spectrum of disorders featured by body's immune responses being directed against its own tissues, resulting in chronic inflammation and tissue damage. Sinomenine (SIN) is an alkaloid isolated from the root and stem of Sinomenium acutum which is mainly used to treat pain, inflammation and immune disorders for centuries in China. Its potential anti-inflammatory role for treating immune-related disorders in experimental animal models and in some clinical applications have been reported widely, suggesting an inspiring application prospect of SIN. In this review, the pharmacokinetics, drug delivery systems, pharmacological mechanisms of action underlying the anti-inflammatory and immunomodulatory effects of SIN, and the possibility of SIN as adjuvant to disease-modifying anti-rheumatic drugs (DMARDs) therapy were summarized and evaluated. This paper aims to reveal the potential prospects and limitations of SIN in the treatment of inflammatory and immune diseases, and to provide ideas for compensating its limitations and reducing the side effects, and thus to make SIN better translate to the clinic.
Collapse
Affiliation(s)
- Siwei Wang
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China; Honghu Hospital of Traditional Chinese Medicine, Honghu 433299, Hubei Province, China
| | - Lvzhuo Zhang
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Yanhua Zhou
- Honghu Hospital of Traditional Chinese Medicine, Honghu 433299, Hubei Province, China
| | - Jiangrong Huang
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China; Jingzhou Central Hospital Affiliated to Yangtze University, Jingzhou 434020, Hubei Province, China.
| | - Zushan Zhou
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China; Honghu Hospital of Traditional Chinese Medicine, Honghu 433299, Hubei Province, China.
| | - Zhenzhen Liu
- Medical Department, Yangtze University, Jingzhou 434023, Hubei Province, China.
| |
Collapse
|
18
|
Fu C, Xin H, Qian Z, Li X, Gao J, Fan Y, Tang Y, Shi Y, Li D, Wu H. Sinomenine Protects against Early Brain Injury by Inhibiting Microglial Inflammatory Response via Nrf2-Dependent Pathway after Subarachnoid Hemorrhage. Brain Sci 2023; 13:brainsci13050716. [PMID: 37239188 DOI: 10.3390/brainsci13050716] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/14/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
Microglial activation and sustained inflammation plays an important role in the processes of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Sinomenine (SIN) has been demonstrated to have neuroprotective effects in the traumatic brain injury (TBI) model. However, the role of SIN in SAH-induced EBI and its latent mechanisms remain unclear. This study was carried out to explore the role of SIN on SAH-induced EBI and its effects on the microglial inflammatory response following SAH. In this study, a model of SAH in rats was established. Modified neurological severity scores (mNSS), encephaledema, and Nissl staining were employed to determine the effects of SIN. Western blot and immunofluorescence analysis were performed to evaluate nuclear factor erythroid 2-related factor 2 (Nrf2) expression. Nrf2-related downstream proteins, including heme oxygenase-1 (HO-1) and quinine oxidoreductase-1 (NQO-1), were detected with immunohistochemistry analyses and Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR). Microglia activation and associated inflammatory factors, factor-kappa B (NF-κB), interleukin-1β (IL-1β), and interleukin-6 (IL-6), were assessed after SAH. The results showed that SIN administration improved neurobehavior function, and attenuated neural apoptosis and brain edema after SAH. In addition, SIN inhibited microglial action and the subsequent inflammatory response after SAH through the upregulated expression of HO-1 and NQO-1 via activation of the Nrf2 pathway. These results demonstrated that SIN supplementation provided protection against SAH-induced neuronal apoptosis by microglial inflammatory response regulation and possible involvement of the Nrf2 pathway.
Collapse
Affiliation(s)
- Chuanjing Fu
- Department of Neurosurgery, Jiangsu Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Heng Xin
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Zhengting Qian
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiang Li
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Juemin Gao
- Department of Neurosurgery, Jiangsu Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Youwu Fan
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yong Tang
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yan Shi
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ding Li
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Heming Wu
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
19
|
Sinomenine ameliorates adjuvant-induced arthritis by inhibiting the autophagy/NETosis/inflammation axis. Sci Rep 2023; 13:3933. [PMID: 36894604 PMCID: PMC9998614 DOI: 10.1038/s41598-023-30922-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Studies have found that neutrophil extracellular traps (NETs) which are the specific dying form of neutrophil upon activation have fundamental role in the rheumatoid arthritis onset and progression. The purpose of this study was to explore the therapeutic effect of Sinomenine on adjuvant-induced arthritis in mice, and the neutrophil activities regulated by Sinomenine. The rheumatoid arthritis model was established by local injection of adjuvant and the Sinomenine treatment was administered orally for 30 days, during which, arthritic scores were evaluated and the joint diameter was measured to determine disease progression. The joint tissues and serum were acquired for further tests after sacrifice. Cytometric beads assay was performed to measure the concentration of cytokines. For paraffin-embedded ankle tissues, hematoxylin and erosin staining and Safranin O-fast staining were adopted to monitor the tissue changes of joint. In order to analyze the inflammation, NETs and autophagy of neutrophils in vivo, immunohistochemistry assays were applied to detect the protein expression levels in the local joints. To describe the effect brought by Sinomenine on inflammation, autophagy and NETs in vitro, the western blotting and the immunofluorescence assays were performed. The joint symptoms of the adjuvant induced arthritis were alleviated by the Sinomenine treatment significantly in terms of the ankle diameter and scores. The improvement of local histopathology changes and decrease of inflammatory cytokines in the serum also confirmed the efficacy. The expression levels of interleukin-6, P65 and p-P65 in the ankle areas of mice were remarkably reduced by Sinomenine. Compared with the model group, the decreased expression levels of lymphocyte antigen 6 complex and myeloperoxidase in the Sinomenine treating group showed the inhibitory effect of Sinomenine on the neutrophil migration. The expression of protein arginine deiminase type 4 (PAD4), ctrullinated histone H3 (CitH3) and microtubule-associated protein 1 light chain 3B (LC3B) had the similar tendency. Upon activation of lipopolysaccharide (LPS) in vitro, Sinomenine suppressed the phosphorylation of P65, extracellular signal-regulated kinase (ERK) and P38 of neutrophil. Meanwhile, Sinomenine inhibited NETs formation induced by phorbol 12-myristate 13-acetate (PMA), which were demonstrated by the decreased expression of neutrophil elastase (NE), PAD4 and CitH3. Sinomenine also inhibited PMA-induced autophagy in vitro based on the changes of Beclin-1 and LC3B. Sinomenine has good efficacy in treating adjuvant induced arthritis via regulating neutrophil activities. Apart from inhibiting activation of nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, the mechanism includes suppression of NETs formation via autophagy inhibition.
Collapse
|
20
|
Unraveling the molecular mechanism of l-menthol against cervical cancer based on network pharmacology, molecular docking and in vitro analysis. Mol Divers 2023; 27:323-340. [PMID: 35467269 DOI: 10.1007/s11030-022-10429-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 03/30/2022] [Indexed: 02/08/2023]
Abstract
Cervical cancer is a major cause of gynecological related mortalities in developing countries. Cisplatin, a potent chemotherapeutic agent used for treating advanced cervical cancer exhibits side effects and resistance development. The current study was aimed to investigate the repurposing of l-menthol as a potential therapeutic drug against cervical cancer. L-menthol was predicted to be non-toxic with good pharmacokinetic properties based on SwissADME and pkCSM analysis. Subsequently, 543 and 1664 targets of l-menthol and cervical cancer were identified using STITCH, BATMAN-TCM, PharmMapper and CTD databases. STRING and Cytoscape analysis of the merged protein-protein interaction network revealed 107 core targets of l- menthol against cervical cancer. M-CODE identified highly connected clusters between the core targets which through KEGG analysis were found to be enriched in pathways related to apoptosis and adherence junctions. Molecular docking showed that l- menthol targeted E6, E6AP and E7 onco-proteins of HPV that interact and inactivate TP53 and Rb1 in cervical cancer, respectively. Molecular docking also showed good binding affinity of l-menthol toward proteins associated with apoptosis and migration. Molecular dynamics simulation confirmed stability of the docked complexes. In vitro analysis confirmed that l-menthol was cytotoxic towards cervical cancer CaSki cells and altered expression of TP53, Rb1, CDKN1A, E2F1, NFKB1, Akt-1, caspase-3, CDH1 and MMP-2 genes identified through network pharmacology approach. Schematic representation of the work flow depicting the potential of l-menthol to target cervical cancer.
Collapse
|
21
|
Wei Z, Chen J, Zuo F, Guo J, Sun X, Liu D, Liu C. Traditional Chinese Medicine has great potential as candidate drugs for lung cancer: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115748. [PMID: 36162545 DOI: 10.1016/j.jep.2022.115748] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE With high mortality and morbidity rates, lung cancer (LC) has become one of the major threats to human health. The treatment strategies for LC currently face issues, such as drug resistance and body tolerance. Traditional Chinese medicine (TCM) is characterized by novel pharmacological mechanisms, low toxicity, and limited side effects. TCM includes a substantial number of biologically active ingredients, several of which are effective monomeric agents against LC. An increasing number of researchers are focusing their efforts on the discovery of active anti-cancer ingredients in TCM. AIM OF THE REVIEW In this review, we summarized the anti-LC mechanisms of five types of TCM monomeric compounds. Our goal is to provide research ideas for the identification of new prospective medication candidates for the treatment of LC. MATERIALS AND METHODS We collected reports on the anti-LC effects of TCM monomers from web databases, including PubMed, Science Direct, Web of Science, and Europe PubMed Central. Among the keywords used were "lung cancer," "traditional Chinese medicine," "pharmacology," and their combinations thereof. Then, we systematically summarized the anti-LC efficacy and related mechanisms of TCM monomers. RESULTS Based on the available literature, this paper reviewed the therapeutic effects and mechanisms of five types of TCM monomers on LC. The characteristics of TCM monomers include the capabilities to suppress the tumor cell cycle, inhibit proliferation, induce apoptosis, promote autophagy, inhibit tumor cell invasion and metastasis, and enhance efficacy or reduce drug resistance when combined with cytotoxic agents and other methods to arrest the progression of LC and prolong the survival of patients. CONCLUSIONS TCM contains numerous flavonoids, alkaloids, terpenoids, polyphenols, and other active compounds that are effective against LC. Given their chemical structure and pharmacological properties, these monomers are suitable as candidate drugs for the treatment of LC.
Collapse
Affiliation(s)
- Zhicheng Wei
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, PR China.
| | - Jing Chen
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, PR China
| | - Fang Zuo
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Julie Guo
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, PR China
| | - Xiaodong Sun
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, PR China
| | - Deming Liu
- Chongqing Clinical Research Center for Dermatology, Chongqing Key Laboratory of Integrative Dermatology Research, Key Laboratory of External Therapies of Traditional Chinese Medicine in Eczema, Department of Dermatology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400011, PR China.
| | - Conghai Liu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, PR China.
| |
Collapse
|
22
|
Chuang YT, Tang JY, Shiau JP, Yen CY, Chang FR, Yang KH, Hou MF, Farooqi AA, Chang HW. Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products. Cancers (Basel) 2023; 15:318. [PMID: 36612314 PMCID: PMC9818271 DOI: 10.3390/cancers15010318] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer-derived exosomes exhibit sophisticated functions, such as proliferation, apoptosis, migration, resistance, and tumor microenvironment changes. Several clinical drugs modulate these exosome functions, but the impacts of natural products are not well understood. Exosome functions are regulated by exosome processing, such as secretion and assembly. The modulation of these exosome-processing genes can exert the anticancer and precancer effects of cancer-derived exosomes. This review focuses on the cancer-derived exosomal miRNAs that regulate exosome processing, acting on the natural-product-modulating cell functions of cancer cells. However, the role of exosomal processing has been overlooked in several studies of exosomal miRNAs and natural products. In this study, utilizing the bioinformatics database (miRDB), the exosome-processing genes of natural-product-modulated exosomal miRNAs were predicted. Consequently, several natural drugs that modulate exosome processing and exosomal miRNAs and regulate cancer cell functions are described here. This review sheds light on and improves our understanding of the modulating effects of exosomal miRNAs and their potential exosomal processing targets on anticancer treatments based on the use of natural products.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Hsueh-Wei Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
23
|
Hashemi M, Mirdamadi MSA, Talebi Y, Khaniabad N, Banaei G, Daneii P, Gholami S, Ghorbani A, Tavakolpournegari A, Farsani ZM, Zarrabi A, Nabavi N, Zandieh MA, Rashidi M, Taheriazam A, Entezari M, Khan H. Pre-clinical and clinical importance of miR-21 in human cancers: Tumorigenesis, therapy response, delivery approaches and targeting agents. Pharmacol Res 2023; 187:106568. [PMID: 36423787 DOI: 10.1016/j.phrs.2022.106568] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022]
Abstract
The field of non-coding RNA (ncRNA) has made significant progress in understanding the pathogenesis of diseases and has broadened our knowledge towards their targeting, especially in cancer therapy. ncRNAs are a large family of RNAs with microRNAs (miRNAs) being one kind of endogenous RNA which lack encoded proteins. By now, miRNAs have been well-coined in pathogenesis and development of cancer. The current review focuses on the role of miR-21 in cancers and its association with tumor progression. miR-21 has both oncogenic and onco-suppressor functions and most of the experiments are in agreement with the tumor-promoting function of this miRNA. miR-21 primarily decreases PTEN expression to induce PI3K/Akt signaling in cancer progression. Overexpression of miR-21 inhibits apoptosis and is vital for inducing pro-survival autophagy. miR-21 is vital for metabolic reprogramming and can induce glycolysis to enhance tumor progression. miR-21 stimulates EMT mechanisms and increases expression of MMP-2 and MMP-9 thereby elevating tumor metastasis. miR-21 is a target of anti-cancer agents such as curcumin and curcumol and its down-regulation impairs tumor progression. Upregulation of miR-21 results in cancer resistance to chemotherapy and radiotherapy. Increasing evidence has revealed the role of miR-21 as a biomarker as it is present in both the serum and exosomes making them beneficial biomarkers for non-invasive diagnosis of cancer.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Motahare Sadat Ayat Mirdamadi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Yasmin Talebi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Islamic Azad University Central Tehran Branch, Tehran, Iran
| | - Nasrin Khaniabad
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Gooya Banaei
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Pouria Daneii
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Sadaf Gholami
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Amin Ghorbani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alireza Tavakolpournegari
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193 Barcelona, Spain
| | - Zoheir Mohammadian Farsani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Industrial and Environmental Biotechnology, National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
24
|
Yum SJ, Jeong HG, Kim SM. Anti-biofilm effects of sinomenine against Staphylococcus aureus. Food Sci Biotechnol 2023; 32:83-90. [PMID: 36606087 PMCID: PMC9807730 DOI: 10.1007/s10068-022-01174-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is a gram-positive foodborne pathogen capable of forming strong biofilms. This study identified that anti-biofilm natural compound against S. aureus. Sinomenine, a natural compound, showed significantly reduced biofilm formation (31.97-39.86%), but no effect on bacterial growth was observed. The dispersion of preformed biofilms was observed by confocal laser scanning microscopy (CLSM). qRT-PCR revealed that sinomenine treatment significantly up-regulated agrA by 3.8-fold and down-regulated icaA gene by 3.1-fold. These indicate that sinomenine treatment induces biofilm dispersal due to cell-cell adhesion, polysaccharide intercellular adhesin (PIA), and phenol-soluble modulin (PSM) peptides production. Our results suggest that sinomenine can be used as a promising agent for effectively controlling biofilm formation and dispersion, thereby making S. aureus more susceptible to the action of antimicrobial agents.
Collapse
Affiliation(s)
- Su-Jin Yum
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Chungnam National University, Daejeon, 34134 Korea
| | - Hee Gon Jeong
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Chungnam National University, Daejeon, 34134 Korea
| | - Seung Min Kim
- Division of Human Ecology, Korea National Open University, Seoul, 03087 Korea
| |
Collapse
|
25
|
Chen X, Lu C, Duan Y, Huang Y. Recent Advancements in Drug Delivery of Sinomenine, A Disease-Modifying Anti-Rheumatic Drug. Pharmaceutics 2022; 14:pharmaceutics14122820. [PMID: 36559313 PMCID: PMC9781253 DOI: 10.3390/pharmaceutics14122820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Sinomenine (SIN) is a benzyltetrahydroisoquinoline-type alkaloid isolated from the dried plant root and stem of Sinomenium acutum (Thumb.) Rehd.et Wils, which shows potent anti-inflammatory and analgesic effects. As a transforming disease-modifying anti-rheumatic drug, SIN has been used to treat rheumatoid arthritis over twenty-five years in China. In recent years, SIN is also in development for use against other disorders, including colitis, pain, traumatic brain injury, and uveitis. However, its commercial hydrochloride (SIN-HCl) shows low oral bioavailability and certain allergic reactions in patients, due to the release of histamine. Therefore, a large number of pharmaceutical strategies have been explored to address these liabilities, such as prolonging release behaviors, enhancing skin permeation and adsorption for transdermal delivery, targeted SIN delivery using new material or conjugates, and co-amorphous technology. This review discusses these different delivery strategies and approaches employed to overcome the limitations of SIN for its efficient delivery, in order to achieve improved bioavailability and reduced side effects. The potential advantages and limitations of SIN delivery strategies are elaborated along with discussions of potential future SIN drug development strategies.
Collapse
Affiliation(s)
- Xin Chen
- Xiangya International Academy of Translational Medicine, Central South University, Changsha 410013, China
| | - Chengcheng Lu
- Xiangya International Academy of Translational Medicine, Central South University, Changsha 410013, China
| | - Yanwen Duan
- Xiangya International Academy of Translational Medicine, Central South University, Changsha 410013, China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha 410013, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha 410011, China
| | - Yong Huang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha 410013, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha 410011, China
- Correspondence:
| |
Collapse
|
26
|
Liu X, Chen L, Wang T. Overcoming cisplatin resistance of human lung cancer by sinomenine through targeting the miR-200a-3p-GLS axis. J Chemother 2022:1-10. [PMID: 36120905 DOI: 10.1080/1120009x.2022.2111490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Lung cancer, a malignant disease, is one of the leading causes of patient death. Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Currently, chemotherapeutic agents such as cisplatin are widely used against lung cancer. However, development of chemoresistance, which led to poor prognosis and low survival rate greatly limited the clinical applications of cisplatin. Sinomenine (SIN) is a bioactive component of sinomenium acutum. Accumulating evidence revealed SIN exhibits potential anti-tumor activities in various types of cancers. However, the precise molecular mechanisms for the sinomenine-induced anti-cancer effects have not been fully elucidated. Here, we assessed the effects of sinomenine on cisplatin sensitivity in NSCLC cells. The combination of SIN with cisplatin showed synergistically inhibitory effects on lung cancer cells by calculating the combination index (CI value) using the Calcusyn 2.0 software. Moreover, we detected that the glutamine metabolism was significantly suppressed by sinomenine treatments in lung cancer cells. Under low glutamine supply, A549 cells showed less sensitivity to sinomenine treatments. Meanwhile, miR-200a-3p was found to be significantly induced by SIN treatments. We demonstrated a suppressive role of miR-200a-3p on glutamine metabolism. Furthermore, miR-200a-3p was downregulated but the glutamine metabolism was significantly hyperactive in A549 cisplatin resistant cells compared with parental cells. Bioinformatical analysis and luciferase assay demonstrated the glutaminase (GLS), a key enzyme of glutamine metabolism, is the direct target of miR-200a-3p in lung cancer cells. Finally, rescue experiments demonstrated that recovery of GLS in miR-200a-3p overexpressing-cisplatin resistant cells successfully overrode the sinomenine-mediated cisplatin sensitization. In summary, this study revealed a new molecular mechanism for the sinomenine-promoted cisplatin sensitization, contributing to investigating the sinomenine-based therapeutic agents against chemoresistant NSCLC.
Collapse
Affiliation(s)
- Xi Liu
- Department of Thoracic Surgery, People's Liberation Army of China General Hospital, Beijing, P. R. China
| | - Lei Chen
- Department of Thoracic Surgery, People's Liberation Army of China General Hospital, Beijing, P. R. China
| | - Tao Wang
- Department of Thoracic Surgery, People's Liberation Army of China General Hospital, Beijing, P. R. China
| |
Collapse
|
27
|
Mafi A, Rahmati A, Babaei Aghdam Z, Salami R, Salami M, Vakili O, Aghadavod E. Recent insights into the microRNA-dependent modulation of gliomas from pathogenesis to diagnosis and treatment. Cell Mol Biol Lett 2022; 27:65. [PMID: 35922753 PMCID: PMC9347108 DOI: 10.1186/s11658-022-00354-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/22/2022] [Indexed: 11/11/2022] Open
Abstract
Gliomas are the most lethal primary brain tumors in adults. These highly invasive tumors have poor 5-year survival for patients. Gliomas are principally characterized by rapid diffusion as well as high levels of cellular heterogeneity. However, to date, the exact pathogenic mechanisms, contributing to gliomas remain ambiguous. MicroRNAs (miRNAs), as small noncoding RNAs of about 20 nucleotides in length, are known as chief modulators of different biological processes at both transcriptional and posttranscriptional levels. More recently, it has been revealed that these noncoding RNA molecules have essential roles in tumorigenesis and progression of multiple cancers, including gliomas. Interestingly, miRNAs are able to modulate diverse cancer-related processes such as cell proliferation and apoptosis, invasion and migration, differentiation and stemness, angiogenesis, and drug resistance; thus, impaired miRNAs may result in deterioration of gliomas. Additionally, miRNAs can be secreted into cerebrospinal fluid (CSF), as well as the bloodstream, and transported between normal and tumor cells freely or by exosomes, converting them into potential diagnostic and/or prognostic biomarkers for gliomas. They would also be great therapeutic agents, especially if they could cross the blood–brain barrier (BBB). Accordingly, in the current review, the contribution of miRNAs to glioma pathogenesis is first discussed, then their glioma-related diagnostic/prognostic and therapeutic potential is highlighted briefly.
Collapse
Affiliation(s)
- Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Atefe Rahmati
- Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Basic Science, Neyshabur University of Medical Science, Neyshabur, Iran
| | - Zahra Babaei Aghdam
- Imaging Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran. .,Department of Clinical Biochemistry, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
28
|
Ding C, Li Y, Sun Y, Wu Y, Wang F, Liu C, Zhang H, Jiang Y, Zhang D, Song X. Sinomenium acutum: A Comprehensive Review of its Botany, Phytochemistry, Pharmacology and Clinical Application. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1219-1253. [PMID: 35681262 DOI: 10.1142/s0192415x22500501] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sinomenium acutumis the dry stem of Sinomenium acutum (Thunb.) Rehd et Wils. (S. acutum) and Sinomenium acutum(Thunb.) Rehd. et Wils. var. cinereumRehd. et Wils and is mainly distributed in China and Japan. As a traditional Chinese medicine (TCM) for dispelling wind and dampness in China, it is widely distributed and has a long history of drug use. In recent years, with the increase of the incidence of rheumatoid disease, S. acutum has become the focus of research. This paper reviews the literature on the chemical constituents, pharmacological effects, clinical applications and pharmacokinetics and safety of S. acutum from the past 60 years. At present, more than 210 natural compounds have been isolated from S. acutum, including alkaloids, lignans, triterpenoid saponins, steroids, and other structures. Pharmacological activities of S. acutum were mainly reported on anti-inflammatory, analgesic, anti-allergic, immunosuppressive, anti-tumor, liver-protective, anti-oxidative, and other effects, and clinical applications were mainly recorded on rheumatoid arthritis, ankylosing spondylitis, and other diseases. The clinical use of SIN has fewer side effects and more safety; only a small number of gastrointestinal reactions occurred, and the symptoms disappeared after the drug stopped. The purpose of this paper is to lay a foundation and provide reference for the follow-up research and wide application of S. acutum.
Collapse
Affiliation(s)
- Chao Ding
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Yuze Li
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Yu Sun
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Ying Wu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Fengrui Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Chenwang Liu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Huawei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Yi Jiang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Dongdong Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| | - Xiaomei Song
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, P. R. China
| |
Collapse
|
29
|
Mohamed A. Ouf A, Abdelrasheed Allam H, Kamel M, Ragab FA, Abdel-Aziz SA. Design, synthesis, cytotoxic and enzyme inhibitory activities of 1,3,4-oxadiazole and 1,3,4-thiadiazine hybrids against non-small cell lung cancer. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2022.100373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
30
|
Li X, Chen W, Huang L, Zhu M, Zhang H, Si Y, Li H, Luo Q, Yu B. Sinomenine hydrochloride suppresses the stemness of breast cancer stem cells by inhibiting Wnt signaling pathway through down-regulation of WNT10B. Pharmacol Res 2022; 179:106222. [DOI: 10.1016/j.phrs.2022.106222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/29/2022] [Accepted: 04/07/2022] [Indexed: 12/22/2022]
|
31
|
Rheumatoid arthritis drug sinomenine induces apoptosis of cervical tumor cells by targeting thioredoxin reductase in vitro and in vivo. Bioorg Chem 2022; 122:105711. [PMID: 35247807 DOI: 10.1016/j.bioorg.2022.105711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/16/2022] [Accepted: 02/25/2022] [Indexed: 12/19/2022]
Abstract
Overexpression of thioredoxin reductase (TrxR) has been linked to tumorigenesis and phenotypic maintenance of malignant tumors. Thus, targeting TrxR with natural molecules is a promising strategy for developing anticancer drugs. Sinomenine is a naturally occurring alkaloid isolated from Sinomenium acutum. The drug, Zhengqing Fengtongning made from sinomenine, has been universally applied in rheumatoid arthritis treatment in China as well as other Asian countries for decades. Recently, increasing evidence indicates that sinomenine appears to be a promising therapeutic agent against various cancer cells. However, the exact mechanism underlying the anticancer activity of sinomenine remains unclear. In this study, we identified sinomenine as a kind of new inhibitor for TrxR. Pharmacological inhibition of TrxR by sinomenine results in the decrease of thiols content, increases the levels of reactive oxygen species, and finally facilitates oxidative stress-mediated cancer cell apoptosis. It is vital that knockdown in TrxR1 by shRNA can increase cell sensitivity to sinomenine. Treatment with sinomenine in vivo leads to a decrease in TrxR activity and tumor growth, and an increase in apoptosis. Our findings provide a novel action mechanism related to sinomenine and presents an insight on how to develop sinomenine as a chemotherapeutic agent for cancer therapy.
Collapse
|
32
|
Bai X, Ali A, Wang N, Liu Z, Lv Z, Zhang Z, Zhao X, Hao H, Zhang Y, Rahman FU. Inhibition of SREBP-mediated lipid biosynthesis and activation of multiple anticancer mechanisms by platinum complexes: Ascribe possibilities of new antitumor strategies. Eur J Med Chem 2022; 227:113920. [PMID: 34742012 DOI: 10.1016/j.ejmech.2021.113920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 12/31/2022]
Abstract
Cancer is one of the most aggressive diseases with poor prognosis and survival rates. Lipids biogenesis play key role in cancer progression, metastasis and tumor development. Suppression of SREBP-mediated lipid biogenesis pathway has been linked with cancer inhibition. Platinum complexes bearing good anticancer effect and multiple genes activation properties are considered important and increase the chances for development of new platinum-based drugs. In this study, we synthesized pyridine co-ligand functionalized cationic complexes and characterized them using multiple spectroscopic and spectrophotometric methods. Two of these complexes were studied in solid state by single crystal X-ray analysis. The stability of these complexes were measured in solution state using 1H NMR methods. These complexes were further investigated for their anticancer activity against human breast, lung and liver cancer cells. MTT assay showed potential cytotoxic activity in dose-dependent manner and decrease survival rates of cancer cells was observed upon treatment with these complexes. Biological assays results revealed higher cytotoxicity as compared to cisplatin and oxaliplatin. Further we studied C2, C6 and C8 in detailed mechanistic anticancer analyses. Clonogenic assay showed decrease survival of MCF-7, HepG2 and A549 cancer cells treated with C2, C6 and C8 as compared to control cells treated with DMSO. TUNEL assay showed more cell death, these complexes suppressed invasion and migration ability of cancer cells and decreased tumor spheroids formation, thus suggesting a potential role in inhibition of cancer metastasis and cancer stem cells formation. Mechanistically, these complexes inhibited sterol regulatory element-binding protein 1 (SREBP-1) expression in cancer cells in dose-dependent manner and thereby reduced lipid biogenesis to suppress cancer progression. Furthermore, expression level was decreased for the key genes LDLR, FASN and HMGCR, those required for sterol biosynthesis. Taken together, these complexes suppressed cancer cell growth, migration, invasion and spheroids formation by inhibiting SREBP-1 mediated lipid biogenesis pathway.
Collapse
Affiliation(s)
- Xue Bai
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Amjad Ali
- Institute of Integrative Biosciences, CECOS University of IT and Emerging Sciences, Peshawar, KPK, Pakistan; Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, People's Republic of China
| | - Na Wang
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Zongwei Liu
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Zhimin Lv
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Zeqing Zhang
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Xing Zhao
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Huifang Hao
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China; School of Life Sciences, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Yongmin Zhang
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China; Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France.
| | - Faiz-Ur Rahman
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China.
| |
Collapse
|
33
|
Ngo DN, Ngo DH, Nguyen HM, Nguyen TH, Nguyen TT, Vo TS. Growth inhibitory activity of Brassica oleracea var. Alboglabra on human gastric cancer cells. JOURNAL OF REPORTS IN PHARMACEUTICAL SCIENCES 2022. [DOI: 10.4103/jrptps.jrptps_119_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
34
|
Sinomenine Inhibits the Progression of Bladder Cancer Cells by Downregulating LncRNA-HEIH Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4699529. [PMID: 34760016 PMCID: PMC8575624 DOI: 10.1155/2021/4699529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022]
Abstract
Background Sinomenine has been reported to effectively repress the progression of lung cancer and breast cancer. However, the effects of sinomenine in bladder cancer are not well understood. The purpose of this study was to evaluate the effects of sinomenine in bladder cancer. Methods The mRNA expression of HEIH in bladder cancer cells was measured by RT-qPCR. T24 and SW780 cells were treated with sinomenine for 24 hours. Cell viability was detected by the MTT assay. Cell migration and invasion were detected by the transwell assay. Western blotting assay was performed to assess the protein expression of Bcl-2, Bax, and caspase-3. Results Sinomenine significantly suppressed cell viability in T24 and SW780 cells. Moreover, cell migration and invasion were significantly inhibited by sinomenine. Sinomenine accelerated the expression of Bax and caspase-3 but decreased the expression of Bcl-2. HEIH was upregulated in bladder cancer cells compared with normal bladder epithelial cells. Besides this, we noticed that HEIH knockdown blocked cell proliferation, migration, and invasion but facilitated cell apoptosis in bladder cancer cells. Additionally, HEIH reversed the suppression of the progression induced by sinomenine. Conclusion Sinomenine was observed to suppress cell progression of bladder cancer cells by inhibiting HEIH expression. Our findings suggested that the use of sinomenine might be an effective treatment for bladder cancer.
Collapse
|
35
|
Peláez R, Ochoa R, Pariente A, Villanueva-Martínez Á, Pérez-Sala Á, Larráyoz IM. Sterculic Acid Alters Adhesion Molecules Expression and Extracellular Matrix Compounds to Regulate Migration of Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13174370. [PMID: 34503180 PMCID: PMC8431022 DOI: 10.3390/cancers13174370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/11/2022] Open
Abstract
Sterculic acid (SA) is a cyclopropenoid fatty acid isolated from Sterculia foetida seeds. This molecule is a well-known inhibitor of SCD1 enzyme, also known as ∆9-desaturase, which main function is related to lipid metabolism. However, recent studies have demonstrated that it also modifies many other pathways and the underlying gene expression. SCD overexpression, or up-regulated activity, has been associated with tumor aggressiveness and poor prognosis in many cancer types. Scd1 down-regulation, with different inhibitors or molecular strategies, reduces tumor cell survival and cell proliferation, as well as the chemoresistance associated with cancer stem cell presence. However, SA effects over cancer cell migration and extracellular matrix or adhesion molecules have not been described in cancer cells up to now. We used different migration assays and qPCR gene expression analysis to evaluate the effects of SA treatment in cancer cells. The results reveal that SA induces tumoral cell death at high doses, but we also observed that lower SA-treatments induce cell adhesion-migration capacity reduction as a result of modifications in the expression of genes related to integrins and extracellular matrix compounds. Overall, the functional and transcriptomic findings suggest that SA could represent a new inhibitor activity of epithelial to mesenchymal transition.
Collapse
Affiliation(s)
- Rafael Peláez
- Correspondence: (R.P.); (I.M.L.); Tel.: +34-941-278-770 ((ext. 84866) (R.P.) & (ext. 89878) (I.M.L.))
| | | | | | | | | | - Ignacio M. Larráyoz
- Correspondence: (R.P.); (I.M.L.); Tel.: +34-941-278-770 ((ext. 84866) (R.P.) & (ext. 89878) (I.M.L.))
| |
Collapse
|
36
|
Yang L, Wang Z. Natural Products, Alone or in Combination with FDA-Approved Drugs, to Treat COVID-19 and Lung Cancer. Biomedicines 2021; 9:689. [PMID: 34207313 PMCID: PMC8234041 DOI: 10.3390/biomedicines9060689] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
As a public health emergency of international concern, the highly contagious coronavirus disease 2019 (COVID-19) pandemic has been identified as a severe threat to the lives of billions of individuals. Lung cancer, a malignant tumor with the highest mortality rate, has brought significant challenges to both human health and economic development. Natural products may play a pivotal role in treating lung diseases. We reviewed published studies relating to natural products, used alone or in combination with US Food and Drug Administration-approved drugs, active against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and lung cancer from 1 January 2020 to 31 May 2021. A wide range of natural products can be considered promising anti-COVID-19 or anti-lung cancer agents have gained widespread attention, including natural products as monotherapy for the treatment of SARS-CoV-2 (ginkgolic acid, shiraiachrome A, resveratrol, and baicalein) or lung cancer (daurisoline, graveospene A, deguelin, and erianin) or in combination with FDA-approved anti-SARS-CoV-2 agents (cepharanthine plus nelfinavir, linoleic acid plus remdesivir) and anti-lung cancer agents (curcumin and cisplatin, celastrol and gefitinib). Natural products have demonstrated potential value and with the assistance of nanotechnology, combination drug therapies, and the codrug strategy, this "natural remedy" could serve as a starting point for further drug development in treating these lung diseases.
Collapse
Affiliation(s)
- Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, China;
| | - Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
37
|
Small in Size, but Large in Action: microRNAs as Potential Modulators of PTEN in Breast and Lung Cancers. Biomolecules 2021; 11:biom11020304. [PMID: 33670518 PMCID: PMC7922700 DOI: 10.3390/biom11020304] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are well-known regulators of biological mechanisms with a small size of 19–24 nucleotides and a single-stranded structure. miRNA dysregulation occurs in cancer progression. miRNAs can function as tumor-suppressing or tumor-promoting factors in cancer via regulating molecular pathways. Breast and lung cancers are two malignant thoracic tumors in which the abnormal expression of miRNAs plays a significant role in their development. Phosphatase and tensin homolog (PTEN) is a tumor-suppressor factor that is capable of suppressing the growth, viability, and metastasis of cancer cells via downregulating phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling. PTEN downregulation occurs in lung and breast cancers to promote PI3K/Akt expression, leading to uncontrolled proliferation, metastasis, and their resistance to chemotherapy and radiotherapy. miRNAs as upstream mediators of PTEN can dually induce/inhibit PTEN signaling in affecting the malignant behavior of lung and breast cancer cells. Furthermore, long non-coding RNAs and circular RNAs can regulate the miRNA/PTEN axis in lung and breast cancer cells. It seems that anti-tumor compounds such as baicalein, propofol, and curcumin can induce PTEN upregulation by affecting miRNAs in suppressing breast and lung cancer progression. These topics are discussed in the current review with a focus on molecular pathways.
Collapse
|
38
|
Qu X, Yu B, Zhu M, Li X, Ma L, Liu C, Zhang Y, Cheng Z. Sinomenine Inhibits the Growth of Ovarian Cancer Cells Through the Suppression of Mitosis by Down-Regulating the Expression and the Activity of CDK1. Onco Targets Ther 2021; 14:823-834. [PMID: 33574676 PMCID: PMC7873025 DOI: 10.2147/ott.s284261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction Ovarian cancer is one of the most common gynecological cancers worldwide. While, therapies against ovarian cancer have not been completely effective, sinomenine has been proved to have anti-tumor activity in various cancer cells. However, study of its anti-ovarian cancer effect is still rare, and the underlying mechanism has not been elucidated. Therefore, we aim to explore the mechanism of sinomenine anti-ovarian cancer. Materials and Methods The effect of anti-ovarian cancer HeyA8 cells was analyzed by CCK8 and colony formation assay. The mechanism of sinomenine anti-ovarian cancer was explored via high throughput RNA-seq, and then the target mRNA and protein expression were verified by real-time PCR and Western blot, respectively. Results We found that the proliferation and clone formation ability of ovarian cancer HeyA8 cells were markedly reduced by 1.56 mM sinomenine. The transcriptome analysis showed that 2679 genes were differentially expressed after sinomenine treatment in HeyA8 cells, including 1323 down-regulated genes and 1356 up-regulated genes. Gene ontology and KEGG pathway enrichment indicated that differential expression genes (DEGs) between the groups of sinomenine and DMSO-treated HeyA8 cells were mainly involved in the process of the cell cycle, such as kinetochore organization, chromosome segregation, and DNA replication. Strikingly, the top 18 ranked degree genes in the protein-protein interaction (PPI) network were mainly involved in the process of mitosis, such as sister chromatid segregation, condensed chromosome, and microtubule cytoskeleton organization. Moreover, real-time PCR results showed consistent expression trends of DEGs with transcriptome analysis. The results of Western blot showed the expression level of CDK1, which was the highest degree gene in PPI and the main regulator controlling the process of mitosis, and the levels of phosphorylated P-CDK (Thr161) and P-Histone H3 (Ser10) were decreased after being treated with sinomenine. Conclusion Our results demonstrated that sinomenine inhibited the proliferation of HeyA8 cells through suppressing mitosis by down-regulating the expression and the activity of CDK1. The study may provide a preliminary research basis for the application of sinomenine in anti-ovarian cancer.
Collapse
Affiliation(s)
- Xiaoyan Qu
- Department of Gynecology and Obstetrics, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Bing Yu
- Department of Cell Biology, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Mengmei Zhu
- Department of Cell Biology, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Xiaomei Li
- Department of Cell Biology, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China.,Cancer Research Laboratory, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Lishan Ma
- Department of Gynecology and Obstetrics, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Chuyin Liu
- Department of Gynecology and Obstetrics, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Yixing Zhang
- Department of Gynecology and Obstetrics, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Zhongping Cheng
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| |
Collapse
|
39
|
Abd-Elaziz K, Jesenak M, Vasakova M, Diamant Z. Revisiting matrix metalloproteinase 12: its role in pathophysiology of asthma and related pulmonary diseases. Curr Opin Pulm Med 2021; 27:54-60. [PMID: 33065600 DOI: 10.1097/mcp.0000000000000743] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Matrix metalloproteinases (MMPs) are a family of over 20 zinc-dependent proteases with different biological and pathological activities, and many have been implicated in several diseases. Although nonselective MMP inhibitors are known to induce serious side-effects, targeting individual MMPs may offer a safer therapeutic potential for several diseases. Hence, we provide a concise overview on MMP-12, given its association with pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis, and other progressive pulmonary fibrosis (PPF), which may also occur in coronavirus disease 2019. RECENT FINDINGS In asthma, COPD, and PPF, increased MMP-12 levels have been associated with inflammation and/or structural changes within the lungs and negatively correlated with functional parameters. Increased pulmonary MMP-12 levels and MMP-12 gene expression have been related to disease severity in asthma and COPD. Targeting MMP-12 showed potential in animal models of pulmonary diseases but human data are still very scarce. SUMMARY Although there may be a potential role of MMP-12 in asthma, COPD and PPF, several pathophysiological aspects await elucidation. Targeting MMP-12 may provide further insights into MMP-12 related mechanisms and how this translates into clinical outcomes; this warrants further research.
Collapse
Affiliation(s)
- Khalid Abd-Elaziz
- Department of Clinical Pharmacology, QPS-Netherlands, Groningen, The Netherlands
| | - Milos Jesenak
- Department of Pediatrics
- Department of Pulmonology and Physiology
- Department of Clinical Immunology and Allergology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Martin, Slovakia
| | - Martina Vasakova
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Zuzana Diamant
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Dept of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|