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
|
Cheng Y, Zhang W, Sun Q, Wang X, Shang Q, Liu J, Zhang Y, Liu R, Sun C. Probing the biological efficacy and mechanistic pathways of natural compounds in breast cancer therapy via the Hedgehog signaling pathway. J Pharm Anal 2025; 15:101143. [PMID: 40291019 PMCID: PMC12023894 DOI: 10.1016/j.jpha.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BC) is one of the most prevalent malignant tumors affecting women worldwide, with its incidence rate continuously increasing. As a result, treatment strategies for this disease have received considerable attention. Research has highlighted the crucial role of the Hedgehog (Hh) signaling pathway in the initiation and progression of BC, particularly in promoting tumor growth and metastasis. Therefore, molecular targets within this pathway represent promising opportunities for the development of novel BC therapies. This study aims to elucidate the therapeutic mechanisms by which natural compounds modulate the Hh signaling pathway in BC. By conducting a comprehensive review of various natural compounds, including polyphenols, terpenes, and alkaloids, we reveal both common and unique regulatory mechanisms that influence this pathway. This investigation represents the first comprehensive analysis of five distinct mechanisms through which natural compounds modulate key molecules within the Hh pathway and their impact on the aggressive behaviors of BC. Furthermore, by exploring the structure-activity relationships between these compounds and their molecular targets, we shed light on the specific structural features that enable natural compounds to interact with various components of the Hh pathway. These novel insights contribute to advancing the development and clinical application of natural compound-based therapeutics. Our thorough review not only lays the groundwork for exploring innovative BC treatments but also opens new avenues for leveraging natural compounds in cancer therapy.
Collapse
Affiliation(s)
- Yining Cheng
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
| | - Qihang Shang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jingyang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China
| | - Yubao Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Ruijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, 261000, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong, 261053, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong, 261000, China
| |
Collapse
|
3
|
Liu R, Yu Y, Wang Q, Zhao Q, Yao Y, Sun M, Zhuang J, Sun C, Qi Y. Interactions between hedgehog signaling pathway and the complex tumor microenvironment in breast cancer: current knowledge and therapeutic promises. Cell Commun Signal 2024; 22:432. [PMID: 39252010 PMCID: PMC11382420 DOI: 10.1186/s12964-024-01812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/31/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer ranks as one of the most common malignancies among women, with its prognosis and therapeutic efficacy heavily influenced by factors associated with the tumor cell biology, particularly the tumor microenvironment (TME). The diverse elements of the TME are engaged in dynamic bidirectional signaling interactions with various pathways, which together dictate the growth, invasiveness, and metastatic potential of breast cancer. The Hedgehog (Hh) signaling pathway, first identified in Drosophila, has been established as playing a critical role in human development and disease. Notably, the dysregulation of the Hh pathway is recognized as a major driver in the initiation, progression, and metastasis of breast cancer. Consequently, elucidating the mechanisms by which the Hh pathway interacts with the distinct components of the breast cancer TME is essential for comprehensively evaluating the link between Hh pathway activation and breast cancer risk. This understanding is also imperative for devising novel targeted therapeutic strategies and preventive measures against breast cancer. In this review, we delineate the current understanding of the impact of Hh pathway perturbations on the breast cancer TME, including the intricate and complex network of intersecting signaling cascades. Additionally, we focus on the therapeutic promise and clinical challenges of Hh pathway inhibitors that target the TME, providing insights into their potential clinical utility and the obstacles that must be overcome to harness their full therapeutic potential.
Collapse
Affiliation(s)
- Ruijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China
| | - Yang Yu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Qingyang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qianxiang Zhao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China
| | - Mengxuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
| | - Yuanfu Qi
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
4
|
Jiang S, Li S, Pang S, Liu M, Sun H, Zhang N, Liu J. A systematic review: Sinomenine. Heliyon 2024; 10:e29976. [PMID: 38765107 PMCID: PMC11098800 DOI: 10.1016/j.heliyon.2024.e29976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/21/2024] Open
Abstract
Sinomenine (SIN), an alkaloid derived from the traditional Chinese medicine, Caulis Sinomenii, has been used as an anti-inflammatory drug in China for over 30 years. With the continuous increase in research on the pharmacological mechanism of SIN, it has been found that, in addition to the typical rheumatoid arthritis (RA) treatment, SIN can be used as a potentially effective therapeutic drug for anti-tumour, anti-renal, and anti-nervous system diseases. By reviewing a large amount of literature and conducting a summary analysis of the literature pertaining to the pharmacological mechanism of SIN, we completed a review that focused on SIN, found that the current research is insufficient, and offered an outlook for future SIN development. We hope that this review will increase the public understanding of the pharmacological mechanisms of SIN, discover SIN research trial shortcomings, and promote the effective treatment of immune diseases, inflammation, and other related diseases.
Collapse
Affiliation(s)
- Shan Jiang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin City, Heilongjiang Province, 150040, PR China
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, 418000, PR China
| | - Shuang Li
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, 418000, PR China
- College Pharmacy, Jiamusi University, Jiamusi City, Heilongjiang Province, 154000, PR China
| | - Siyuan Pang
- Hunan Zhengqing Pharmaceutical Company Group Ltd, Huaihua City, Hunan Province, 418000, PR China
| | - Mei Liu
- School of Pharmaceutical Sciences, University of South China, Hengyang City, Hunan Province, 421001, PR China
| | - Huifeng Sun
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin City, Heilongjiang Province, 150040, PR China
| | - Ning Zhang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin City, Heilongjiang Province, 150040, PR China
| | - Jianxin Liu
- Sino-Pakistan Center on Traditional Chinese Medicine, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua City, Hunan Province, 418000, PR China
- School of Pharmaceutical Sciences, University of South China, Hengyang City, Hunan Province, 421001, PR China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha City, Hunan Province, 410208, PR China
| |
Collapse
|
5
|
Huang A, Xu T, Lu X, Ma L, Ma H, Yu Y, Yao L. Shh-Gli2-Runx2 inhibits vascular calcification. Nephrol Dial Transplant 2024; 39:305-316. [PMID: 37451818 DOI: 10.1093/ndt/gfad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND In patients with chronic kidney disease (CKD), vascular calcification (VC) is common and is associated with a higher risk of all-cause mortality. Shh, one ligand for Hedgehog (Hh) signaling, participates in osteogenesis and several cardiovascular diseases. However, it remains unclear whether Shh is implicated in the development of VC. METHODS Inorganic phosphorus 2.6 mM was used to induce vascular smooth muscle cells (VSMCs) calcification. Mice were fed with adenine diet supplement with 1.2% phosphorus to induce VC. RESULTS Shh was decreased in VSMCs exposed to inorganic phosphorus, calcified arteries in mice fed with an adenine diet, as well as radial arteries from patients with CKD presenting VC. Overexpression of Shh inhibited VSMCs ostosteoblastic differentiation and calcification, whereas its silencing accelerated these processes. Likewise, mice treated with smoothened agonist (SAG; Hh signaling agonist) showed alleviated VC, and mice treated with cyclopamine (CPN; Hh signaling antagonist) exhibited severe VC. Additionally, overexpression of Gli2 significantly reversed the pro-calcification effect of Shh silencing on VSMCs, suggesting that Shh inhibited VC via Gli2. Mechanistically, Gli2 interacted with Runx2 and promoted its ubiquitin proteasomal degradation, therefore protecting against VC. Of interest, the pro-degradation effect of Gli2 on Runx2 was independent of Smurf1 and Cullin4B. CONCLUSIONS Our study provided deeper insight to the pathogenesis of VC, and Shh might be a novel potential target for VC treatment.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ling Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Haiying Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Shenyang Engineering Technology R&D Center of Cell Therapy Co. Ltd, Shenyang, China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Hou W, Huang L, Huang H, Liu S, Dai W, Tang J, Chen X, Lu X, Zheng Q, Zhou Z, Zhang Z, Lan J. Bioactivities and Mechanisms of Action of Sinomenine and Its Derivatives: A Comprehensive Review. Molecules 2024; 29:540. [PMID: 38276618 PMCID: PMC10818773 DOI: 10.3390/molecules29020540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Sinomenine, an isoquinoline alkaloid extracted from the roots and stems of Sinomenium acutum, has been extensively studied for its derivatives as bioactive agents. This review concentrates on the research advancements in the biological activities and action mechanisms of sinomenine-related compounds until November 2023. The findings indicate a broad spectrum of pharmacological effects, including antitumor, anti-inflammation, neuroprotection, and immunosuppressive properties. These compounds are notably effective against breast, lung, liver, and prostate cancers, exhibiting IC50 values of approximately 121.4 nM against PC-3 and DU-145 cells, primarily through the PI3K/Akt/mTOR, NF-κB, MAPK, and JAK/STAT signaling pathways. Additionally, they manifest anti-inflammatory and analgesic effects predominantly via the NF-κB, MAPK, and Nrf2 signaling pathways. Utilized in treating rheumatic arthritis, these alkaloids also play a significant role in cardiovascular and cerebrovascular protection, as well as organ protection through the NF-κB, Nrf2, MAPK, and PI3K/Akt/mTOR signaling pathways. This review concludes with perspectives and insights on this topic, highlighting the potential of sinomenine-related compounds in clinical applications and the development of medications derived from natural products.
Collapse
Affiliation(s)
- Wen Hou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Lejun Huang
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China;
| | - Hao Huang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Shenglan Liu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Wei Dai
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jianhong Tang
- Laboratory Animal Engineering Research Center of Ganzhou, Gannan Medical University, Ganzhou 341000, China;
| | - Xiangzhao Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Xiaolu Lu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Qisheng Zheng
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Zhinuo Zhou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Ziyun Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jinxia Lan
- College of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
7
|
Keita A, Duval R, Porée FH. Chemistry and biology of ent-morphinan alkaloids. THE ALKALOIDS. CHEMISTRY AND BIOLOGY 2023; 90:1-96. [PMID: 37716795 DOI: 10.1016/bs.alkal.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
Abstract
Morphinan alkaloids have attracted constant attention since the isolation of morphine by Sertürner in 1805. However, a group of 45 compounds possessing a complete ent-morphinan backbone can also be found in the literature. These compounds are related to the morphinandienone subgroup and display a substitution pattern which is different from the morphinans. In particular, these alkaloids could be substituted at position C-2 and C-8 either by a hydroxy function or a methoxy moiety. Four groups of ent-morphinan alkaloids can be proposed, the salutaridine, pallidine, cephasugine and erromangine series. Interestingly, the botanical distribution of the ent-morphinans is more widespread than for the morphinans and includes the Annonaceae, Berberidaceae, Euphorbiaceae, Fumariaceae, Hernandiaceae, Lauraceae, Menispermaceae, Monimiaceae, Papaveraceae, and Ranunculaceae families. To date, their exact mode of production remains elusive and their interplay with the biosynthetic pathway of other classes of benzyltetrahydroisoquinoline alkaloids, in particular aporphines, should be confirmed. Exploration of the biological and therapeutic potential of these compounds is limited to some areas, namely central nervous system (CNS), inflammation, cancer, malaria and viruses. Further studies should be conducted to identify the cellular/molecular targets in view of promoting these compounds as new scaffolds in medicinal chemistry.
Collapse
Affiliation(s)
| | - Romain Duval
- Université Paris Cité, IRD, MERIT, Paris, France.
| | | |
Collapse
|
8
|
Ma C, Hu K, Ullah I, Zheng QK, Zhang N, Sun ZG. Molecular Mechanisms Involving the Sonic Hedgehog Pathway in Lung Cancer Therapy: Recent Advances. Front Oncol 2022; 12:729088. [PMID: 35433472 PMCID: PMC9010822 DOI: 10.3389/fonc.2022.729088] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 03/03/2022] [Indexed: 12/09/2022] Open
Abstract
According to the latest statistics from the International Agency for Research on Cancer (IARC), lung cancer is one of the most lethal malignancies in the world, accounting for approximately 18% of all cancer-associated deaths. Yet, even with aggressive interventions for advanced lung cancer, the five-year survival rate remains low, at around 15%. The hedgehog signaling pathway is highly conserved during embryonic development and is involved in tissue homeostasis as well as organ development. However, studies have documented an increasing prevalence of aberrant activation of HH signaling in lung cancer patients, promoting malignant lung cancer progression with poor prognostic outcomes. Inhibitors targeting the HH pathway have been widely used in tumor therapy, however, they still cannot avoid the occurrence of drug resistance. Interestingly, natural products, either alone or in combination with chemotherapy, have greatly improved overall survival outcomes for lung cancer patients by acting on the HH signaling pathway because of its unique and excellent pharmacological properties. In this review, we elucidate on the underlying molecular mechanisms through which the HH pathway promotes malignant biological behaviors in lung cancer, as well as the potential of inhibitors or natural compounds in targeting HH signaling for clinical applications in lung cancer therapy.
Collapse
Affiliation(s)
- Chao Ma
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Kang Hu
- School of Clinical Medicine, Weifang Medical University, Weifang, China
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Irfan Ullah
- Department of Surgery, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Qing-Kang Zheng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Nan Zhang
- Breast Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
9
|
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]
|
10
|
Song L, Tang L, Lu D, Hu M, Liu C, Zhang H, Zhao Y, Liu D, Zhang S. Sinomenine Inhibits Vasculogenic Mimicry and Migration of Breast Cancer Side Population Cells via Regulating miR-340-5p/SIAH2 Axis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4914005. [PMID: 35309179 PMCID: PMC8926463 DOI: 10.1155/2022/4914005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/29/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia and its induced vasculogenic mimicry (VM) formation, which both closely related with stem-like side population (SP) cells, are the main culprits leading to tumor invasion and metastasis. Sinomenine exhibits excellent anticancer activity in breast cancer, but whether and how it affects hypoxia-triggered VM formation in breast cancer SP cells remains unclear. In this study, breast cancer SP cells were sorted from MDA-MB-231 cells and cultured with sinomenine under hypoxic conditions. Sinomenine obviously repressed the migration and VM formation of breast cancer SP cells. Through downregulating SIAH2 and HIF-1α, sinomenine can inhibit epithelial-mesenchymal transition process of breast cancer SP cells. SIAH2 was identified as a target of miR-340-5p and was downregulated by it, and sinomenine can upregulate miR-340-5p. Hypoxia-induced downregulation of miR-340-5p and activation of SIAH2/HIF-1α pathway can be both counteracted by the sinomenine. Moreover, miR-340-5p inhibition and SIAH2 overexpression can partly counteract the anticancer effects of sinomenine. Taken together, sinomenine inhibits hypoxia-caused VM formation and metastasis of breast cancer SP cells by regulating the miR-340-5p/SIAH2 axis.
Collapse
Affiliation(s)
- Lingqin Song
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China
| | - Liqiong Tang
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China
| | - Dalin Lu
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, 510632 Guangdong, China
| | - Min Hu
- Department of Applied Chemistry, School of Science, Xi'an Jiaotong University, Xi'an, 710049 Shaanxi, China
| | - Chengcheng Liu
- Department of Pathogenic Microbiology & Immunology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061 Shaanxi, China
| | - Haifeng Zhang
- Department of Pathology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061 Shaanxi, China
| | - Yang Zhao
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China
| | - Di Liu
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China
| | - Shuqun Zhang
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China
| |
Collapse
|
11
|
Zhao W, Liu J, Li Y, Chen Z, Qi D, Zhang Z. Immune Effect of Active Components of Traditional Chinese Medicine on Triple-Negative Breast Cancer. Front Pharmacol 2021; 12:731741. [PMID: 34925002 PMCID: PMC8678494 DOI: 10.3389/fphar.2021.731741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancers are heterogeneous, poorly prognostic, and metastatic malignancies that result in a high risk of death for patients. Targeted therapy for triple-negative breast cancer has been extremely challenging due to the lack of expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Clinical treatment regimens for triple-negative breast cancer are often based on paclitaxel and platinum drugs, but drug resistance and side effects from the drugs frequently lead to treatment failure, thus requiring the development of new therapeutic platforms. In recent years, research on traditional Chinese medicine in modulating the immune function of the body has shown that it has the potential to be an effective treatment option against triple-negative breast cancer. Active components of herbal medicines such as alkaloids, flavonoids, polyphenols, saponins, and polysaccharides have been shown to inhibit cancer cell proliferation and metastasis by activating inflammatory immune responses and can modulate tumor-related signaling pathways to further inhibit the invasion of triple-negative breast cancer. This paper reviews the immunomodulatory mechanisms of different herbal active ingredients against triple-negative breast cancer and provides an outlook on the challenges and directions of development for the treatment of triple-negative breast cancer with herbal active ingredients.
Collapse
Affiliation(s)
- Wenjie Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhua Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yaqun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zichao Chen
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongmei Qi
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
12
|
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
|
13
|
Zheng X, Li W, Xu H, Liu J, Ren L, Yang Y, Li S, Wang J, Ji T, Du G. Sinomenine ester derivative inhibits glioblastoma by inducing mitochondria-dependent apoptosis and autophagy by PI3K/AKT/mTOR and AMPK/mTOR pathway. Acta Pharm Sin B 2021; 11:3465-3480. [PMID: 34900530 PMCID: PMC8642618 DOI: 10.1016/j.apsb.2021.05.027] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/22/2021] [Accepted: 05/19/2021] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) in the central nervous system is the most lethal advanced glioma and currently there is no effective treatment for it. Studies of sinomenine, an alkaloid from the Chinese medicinal plant, Sinomenium acutum, showed that it had inhibitory effects on several kinds of cancer. Here, we synthesized a sinomenine derivative, sino-wcj-33 (SW33), tested it for antitumor activity on GBM and explored the underlying mechanism. SW33 significantly inhibited proliferation and colony formation of GBM and reduced migration and invasion of U87 and U251 cells. It also arrested the cell cycle at G2/M phase and induced mitochondria-dependent apoptosis. Differential gene enrichment analysis and pathway validation showed that SW33 exerted anti-GBM effects by regulating PI3K/AKT and AMPK signaling pathways and significantly suppressed tumorigenicity with no obvious adverse effects on the body. SW33 also induced autophagy through the PI3K/AKT/mTOR and AMPK/mTOR pathways. Thus, SW33 appears to be a promising drug for treating GBM effectively and safely.
Collapse
|
14
|
Yang W, Feng Q, Li M, Su J, Wang P, Wang X, Yin Y, Wang X, Zhao M. Sinomenine Suppresses Development of Hepatocellular Carcinoma Cells via Inhibiting MARCH1 and AMPK/STAT3 Signaling Pathway. Front Mol Biosci 2021; 8:684262. [PMID: 34179090 PMCID: PMC8222788 DOI: 10.3389/fmolb.2021.684262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Promotion of apoptosis and suppression of proliferation in tumor cells are popular strategies for developing anticancer drugs. Sinomenine (SIN), a plant-derived alkaloid, displays antitumor activity. However, the mechanism of action of SIN against hepatocellular carcinoma (HCC) is unclear. Herein, several molecular technologies, such as Western Blotting, qRT-PCR, flow cytometry, and gene knockdown were applied to explore the role and mechanism of action of SIN in the treatment of HCC. It was found that SIN arrests HCC cell cycle at G0/G1 phase, induces apoptosis, and suppresses proliferation of HCC cells via down-regulating the expression of membrane-associated RING-CH finger protein 1 (MARCH1). Moreover, SIN induces cell death and growth inhibition through AMPK/STAT3 signaling pathway. MARCH1 expression was silenced by siRNA to explore its involvement in the regulation of AMPK/STAT3 signaling pathway. Silencing MARCH1 caused down-regulation of phosphorylation of AMPK, STAT3 and decreased cell viability and function. Our results suggested that SIN inhibits proliferation and promotes apoptosis of HCC cells by MARCH1-mediated AMPK/STAT3 signaling pathway. This study provides new support for SIN as a clinical anticancer drug and illustrates that targeting MARCH1 could be a novel treatment strategy in developing anticancer therapeutics.
Collapse
Affiliation(s)
- Wei Yang
- Department of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Qihua Feng
- Department of Imaging, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Minjing Li
- Department of Chinese Medicine Prescription, Binzhou Medical University, Yantai, China
| | - Jiaqi Su
- Department of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Peiyuan Wang
- Department of Imaging, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xu Wang
- Department of Imaging, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yancun Yin
- Department of Human Anatomy, Binzhou Medical University, Yantai, China
| | - Xia Wang
- Department of Oral Pathology, Binzhou Medical University, Yantai, China
| | - Mingdong Zhao
- Department of Medical Imaging, Binzhou Medical University, Yantai, China
| |
Collapse
|
15
|
García-Martínez A, Pérez-Balaguer A, Ortiz-Martínez F, Pomares-Navarro E, Sanmartín E, García-Escolano M, Montoyo-Pujol YG, Castellón-Molla E, Peiró G. Hedgehog gene expression patterns among intrinsic subtypes of breast cancer: Prognostic relevance. Pathol Res Pract 2021; 223:153478. [PMID: 34022683 DOI: 10.1016/j.prp.2021.153478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Hedgehog (Hh) signaling is a crucial developmental regulatory pathway recognized as a primary oncogenesis driver in various human cancers. However, its role in breast carcinoma (BC) has been underexplored. METHODS We analyzed the expression of several Hh associated genes in a clinical series and breast cancer cell lines. We included 193 BC stratified according to intrinsic immunophenotypes. Gene expression profiling ofBOC, PTCH, SMO, GLI1, GLI2, and GLI3 was performed by qRT-PCR. Results were correlated with clinical-pathological variables and outcome. RESULTS We observed expression ofGLI2 in triple-negative/basal-like (TN/BL) and GLI3 in luminal cells. In samples, BOC, GLI1, GLI2, and GLI3 expression correlated significantly with luminal tumors and good prognostic factors. In contrast, PTCH and SMO correlated with TN/BL phenotype and nodal involvement. Patients whose tumors expressed SMO had a poorer outcome, especially those with HER2 phenotype. Positive lymph-node status and high SMO remained independent poor prognostic factors. CONCLUSION Our results support a differential Hh pathway activation in BC phenotypes.SMO levels stratified patients at risk of recurrence and death in HER2 phenotype, and it showed an independent prognostic value. Therefore, SMO could be a potential therapeutic target for a subset of BC patients.
Collapse
Affiliation(s)
- Araceli García-Martínez
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain.
| | - Ariadna Pérez-Balaguer
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Fernando Ortiz-Martínez
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Eloy Pomares-Navarro
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Elena Sanmartín
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Marta García-Escolano
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Yoel G Montoyo-Pujol
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Elena Castellón-Molla
- Pathology Dept., University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| | - Gloria Peiró
- Research Unit, University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain; Pathology Dept., University General Hospital of Alicante, and Alicante Institute for Health and Biomedical Research (ISABIAL), Pintor Baeza 12, 03010 Alicante, Spain
| |
Collapse
|
16
|
Li XM, Li MT, Jiang N, Si YC, Zhu MM, Wu QY, Shi DC, Shi H, Luo Q, Yu B. Network Pharmacology-Based Approach to Investigate the Molecular Targets of Sinomenine for Treating Breast Cancer. Cancer Manag Res 2021; 13:1189-1204. [PMID: 33603465 PMCID: PMC7881794 DOI: 10.2147/cmar.s282684] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Sinomenine has been known to inhibit the proliferation of breast cancer cells. However, its targets have not been found yet. This study aimed to search for molecular targets of sinomenine for treating breast cancer via network pharmacology. Methods Potential targets of sinomenine or breast cancer were separately screened from indicated databases. The common targets of both sinomenine and breast cancer were considered as the targets of sinomenine for treating breast cancer. A sinomenine-target-pathway network was constructed based on the obtained results from Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The putative targets of sinomenine were further determined by using protein–protein interaction (PPI) analysis and molecular docking. Finally, the putative targets were verified in vitro and in vivo. Results Twenty predicted targets were identified through network pharmacological analysis. Gene Ontology (GO) and KEGG pathway enrichment indicated that these predicted targets enriched in the process of MAP kinase activity, VEGF signaling pathway, Relaxin signaling pathway, Growth hormone synthesis, secretion and action. MAPK1, NOS3, NR3C1, NOS1 and NOS2 were further identified as the putative targets by using PPI and molecular docking analysis. Expression of MAPK1, NR3C1, NOS1, NOS2 and NOS3 genes were significantly regulated by sinomenine in both MCF-7 cells and MDA-MB-231 cells. Furthermore, the expression of NR3C1 in human breast cancer specimens was lower than that in para-tumor normal tissues. Meanwhile, the expression of NR3C1 in xenograft tumors was up-regulated after sinomenine treatment. Conclusion MAPK1, NR3C1, NOS1, NOS2 and NOS3 were identified as the putative targets of sinomenine for treating breast cancer. NR3C1 was preliminarily confirmed as a target of sinomenine in two breast cancer cell lines, xenograft tumor models and human breast cancer specimens. These data indicated that the network pharmacology-based prediction of sinomenine targets for treating breast cancer could be reliable.
Collapse
Affiliation(s)
- Xiao-Mei Li
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China.,Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Mao-Ting Li
- Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China.,Student Brigade, Second Military Medical University, Shanghai, People's Republic of China
| | - Ni Jiang
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Ya-Chen Si
- Student Brigade, Second Military Medical University, Shanghai, People's Republic of China
| | - Meng-Mei Zhu
- Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Qiao-Yuan Wu
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Dong-Chen Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Shanghai, 200433, People's Republic of China
| | - Hui Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Shanghai, 200433, People's Republic of China
| | - Qing Luo
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Bing Yu
- Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| |
Collapse
|
17
|
Sinomenine inhibits hypoxia induced breast cancer side population cells metastasis by PI3K/Akt/mTOR pathway. Bioorg Med Chem 2020; 31:115986. [PMID: 33412412 DOI: 10.1016/j.bmc.2020.115986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 01/23/2023]
Abstract
Sinomenine is an alkaloid derived from Chinese medicinal plant Sinomenium acutum. Our previous studies suggested that sinomenine can inhibit the metastasis of breast cancer. However, whether sinomenine can inhibit the metastasis characteristics of breast cancer side population (SP) cells is still unknown. In present study, we isolated the side population (SP) cells from MDA-MB-231 cells by fluorescence-activated cell sorting (FACS). MDA-MB-231 SP cells were treated with different concentrations of sinomenine at the absence or presence of hypoxia, and cell viability were measured by CCK-8 assay. The transwell invasive assay were conducted to assess of the effect of sinomenine on the invasion of hypoxic MDA-MB-231 SP cells. The protein expression was detected by Western blot assay. Sinomenine inhibited the cell viability and invasion of hypoxic MDA-MB-231 SP cells. Western blot assay results showed that the upregulation of MMP-2 and MMP-9 by hypoxia was inversed by sinomenine. Additionally, it was found that sinomenine suppressed the activation of PI3K/Akt/mTOR pathway under hypoxia in MDA-MB-231 SP cells. Moreover, the inhibiton of sinomenine on metastasis of hypoxic MDA-MB-231SP cells and PI3K/Akt/mTOR pathway could be rescued by PI3K activator IGF-1. Our study suggested that sinomenine inhibits invasion of breast cancer SP cells under hypoxia through PI3K/Akt/mTOR pathway.
Collapse
|
18
|
Sinomenine sensitizes human gastric cancer cells to cisplatin through negative regulation of PI3K/AKT/Wnt signaling pathway. Anticancer Drugs 2020; 30:983-990. [PMID: 31609766 PMCID: PMC6824511 DOI: 10.1097/cad.0000000000000834] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sinomenine (SIN) has been reported its antitumor effects on various types of human cancers, but there is no available information regarding the antitumor effects of SIN and cisplatin on gastric cancer. Here, we examined the antitumor effects of SIN combined with cisplatin on gastric cancer cells as well as the underlying biological mechanisms. CCK-8 assay and Calcusyn 2.0 software analysis, Hoechst 33258 staining and flow cytometry, transwell assay showed that SIN and cisplatin synergistically inhibited growth, induced apoptosis, and suppressed invasion than did either drug alone in gastric cancer cells. Interestingly, no change in the AKT level was found, whereas SIN and cisplatin led to a dramatic decrease in p-AKT level compared with either alone treatment. SIN and cisplatin further decreased the Bcl-2, procaspase-3, and β-catenin, but increased Bax, cleaved dcaspase 3, MMP9, and MMP2 in combined group than in either alone group. Immunofluorescence staining showed again a significant decrease in nucleus β-catenin was found in combined group. These data suggested that SIN sensitizes human gastric cancer cells to cisplatin through negative regulation of PI3K/AKT/Wnt signaling pathway. In conclusion, SIN and cisplatin exerted synergistic antitumor effects in gastric cancer cells and might constitute a promising therapeutic approach for gastric cancer.
Collapse
|
19
|
Gao G, Liang X, Ma W. Sinomenine restrains breast cancer cells proliferation, migration and invasion via modulation of miR-29/PDCD-4 axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3839-3846. [PMID: 31556312 DOI: 10.1080/21691401.2019.1666861] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sinomenine (Sino) is diffusely applied in heal rheumatoid arthritis and neuralgia. Howbeit, the activities of Sino in breast cancer cells remain confused. The research attempted to probe the anti-tumor function of Sino in breast cancer cells and divulge the feasible molecular mechanism. Sion at the 1-16 μM concentrations was exploited for the exposure of MDA-MB-231 or MCF7 cells, and cell growth, migration, invasion, cell cycle-relevant and apoptosis-correlative factors were estimated. Micro RNA (miR)-29 expression was evaluated via enforcing qRT-PCR, and the actions of miR-29 in MDA-MB-231 cells growth, migration and invasion were appraised after the overexpressed or suppressed vectors transfection. The functions of PDCD-4 in JNK and MEK/ERK pathways were estimated by employing western blot. We found that, Sino exposure impeded cell proliferation, provoked cell apoptosis and barricaded cell migration and invasion in MDA-MB-231 and MCF7 cells. Enhancement of miR-29 was observed in Sino-managed cells, and miR-29 overexpression further potentiated the activities of Sino in MDA-MB-231 cells. Additionally, Sino remarkably enhanced PCDC-4 expression via adjusting miR-29 in MDA-MB-231 cells. Beyond that, overexpressed PCDC-4 obstructed JNK and MEK/ERK pathways in MDA-MB-231 cells. Taken together, the explorations unveiled that Sino restrained MDA-MB-231 cells proliferation, migration, invasion, and provoked apoptosis through modulation of miR-29/PDCD-4 axis. Highlight Sino inhibits MDA-MB-231 and MCF7 cells proliferation and provokes apoptosis; Sino restrains MDA-MB-231 and MCF7 cells migration and invasion; Sino ascends miR-29 expression in MDA-MB-231 and MCF7 cells; Sino adjusts cell growth, migration and invasion via modulating miR-29; Sino up-regulates PDCD-4 expression through mediating miR-29; PDCD-4 obstructs JNK and MEK/ERK pathways in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Guanglei Gao
- Department of Galactophore, Linyi Central Hospital , Linyi , China
| | - Xiaolin Liang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University , Shenzhen , China
| | - Wenyan Ma
- Department of Pharmacy, Jining No.1 People's Hospital , Jining , China
| |
Collapse
|
20
|
Efferth T, Oesch F. Repurposing of plant alkaloids for cancer therapy: Pharmacology and toxicology. Semin Cancer Biol 2019; 68:143-163. [PMID: 31883912 DOI: 10.1016/j.semcancer.2019.12.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/15/2019] [Indexed: 02/08/2023]
Abstract
Drug repurposing (or repositioning) is an emerging concept to use old drugs for new treatment indications. Phytochemicals isolated from medicinal plants have been largely neglected in this context, although their pharmacological activities have been well investigated in the past, and they may have considerable potentials for repositioning. A grand number of plant alkaloids inhibit syngeneic or xenograft tumor growth in vivo. Molecular modes of action in cancer cells include induction of cell cycle arrest, intrinsic and extrinsic apoptosis, autophagy, inhibition of angiogenesis and glycolysis, stress and anti-inflammatory responses, regulation of immune functions, cellular differentiation, and inhibition of invasion and metastasis. Numerous underlying signaling processes are affected by plant alkaloids. Furthermore, plant alkaloids suppress carcinogenesis, indicating chemopreventive properties. Some plant alkaloids reveal toxicities such as hepato-, nephro- or genotoxicity, which disqualifies them for repositioning purposes. Others even protect from hepatotoxicity or cardiotoxicity of xenobiotics and established anticancer drugs. The present survey of the published literature clearly demonstrates that plant alkaloids have the potential for repositioning in cancer therapy. Exploitation of the chemical diversity of natural alkaloids may enrich the candidate pool of compounds for cancer chemotherapy and -prevention. Their further preclinical and clinical development should follow the same stringent rules as for any other synthetic drug as well. Prospective randomized, placebo-controlled clinical phase I and II trials should be initiated to unravel the full potential of plant alkaloids for drug repositioning.
Collapse
Affiliation(s)
- Thomas Efferth
- Department of Pharmaceutical Biology, Johannes Gutenberg University, Mainz, Germany.
| | - Franz Oesch
- Institute of Toxicology, Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
21
|
Gao LN, Zhong B, Wang Y. Mechanism Underlying Antitumor Effects of Sinomenine. Chin J Integr Med 2019; 25:873-878. [DOI: 10.1007/s11655-019-3151-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2018] [Indexed: 02/06/2023]
|
22
|
Zhang YS, Han JY, Iqbal O, Liang AH. Research Advances and Prospects on Mechanism of Sinomenin on Histamine Release and the Binding to Histamine Receptors. Int J Mol Sci 2018; 20:ijms20010070. [PMID: 30586944 PMCID: PMC6337707 DOI: 10.3390/ijms20010070] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022] Open
Abstract
Sinomenine (SIN) is widely used in China to treat a variety of rheumatic diseases (RA), and has various pharmacological effects such as anti-inflammatory, analgesic, and anti-tumor effects. However, due to the histamine release characteristics of SIN, its adverse reactions such as allergic reactions, gastrointestinal reactions, and circulatory systemic reactions have been drawing increasing attention. We present here a systematic review of the chemical structure, pharmacological effects, clinical application, and adverse reactions of SIN, a detailed discussion on the relationship between histamine/histamine receptor and mechanism of action of SIN. In addition, we simulated the binding of SIN to four histamine receptors by using a virtual molecular docking method and found that the bonding intensity between SIN and receptors varied in the order shown as follows: H1R > H2R ~ H3R > H4R. The docking results suggested that SIN might exhibit dual regulatory effects in many processes such as cyclooxygenase-2 (COX-2) expression, NF-κB pathway activation, and degranulation of mast cells to release histamine, thereby exhibiting pro-inflammatory (adverse reactions)/anti-inflammatory effects. This study provides a theoretical basis for the clinical treatment of inflammations seen such as in RA using SIN, and also suggests that SIN has great potential in the field of cancer treatment and will have very important social and economic significance.
Collapse
Affiliation(s)
- Yu-Shi Zhang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jia-Yin Han
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Omer Iqbal
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL 60153, USA.
| | - Ai-Hua Liang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
23
|
Xu W, Wang X, Tu Y, Masaki H, Tanaka S, Onda K, Sugiyama K, Yamada H, Hirano T. Plant‐derived alkaloid sinomenine potentiates glucocorticoid pharmacodynamics in mitogen‐activated human peripheral blood mononuclear cells by regulating the translocation of glucocorticoid receptor. Phytother Res 2018; 33:187-196. [DOI: 10.1002/ptr.6215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/05/2018] [Accepted: 09/26/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Wencheng Xu
- Department of Pharmacy Hubei Provincial Hospital of Traditional Chinese Medicine Wuhan China
- Institute of Traditional Chinese Medicine Hubei Province Academy of Traditional Chinese Medicine Wuhan China
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| | - Xiaoqin Wang
- Institute of Traditional Chinese Medicine Hubei Province Academy of Traditional Chinese Medicine Wuhan China
- Department of Nephrology Hubei Provincial Hospital of Traditional Chinese Medicine Wuhan China
| | - Yuanchao Tu
- Institute of Traditional Chinese Medicine Hubei Province Academy of Traditional Chinese Medicine Wuhan China
- Department of Nephrology Hubei Provincial Hospital of Traditional Chinese Medicine Wuhan China
| | - Hiroshi Masaki
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| | - Sachiko Tanaka
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| | - Kenji Onda
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| | - Kentaro Sugiyama
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| | - Haruki Yamada
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| | - Toshihiko Hirano
- Department of Clinical Pharmacology, School of Pharmacy Tokyo University of Pharmacy and Life Sciences Hachioji Japan
| |
Collapse
|
24
|
Jian W, Zhang X, Wang J, Liu Y, Hu C, Wang X, Liu R. Scinderin-knockdown inhibits proliferation and promotes apoptosis in human breast carcinoma cells. Oncol Lett 2018; 16:3207-3214. [PMID: 30127916 DOI: 10.3892/ol.2018.9009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 05/11/2018] [Indexed: 01/16/2023] Open
Abstract
Previous studies have reported that scinderin (SCIN) affects multiple cellular processes, including proliferation, migration and differentiation in cancer. However, the specific role of SCIN in breast cancer (BC) cells is unknown. Immunohistochemistry was used to investigate SCIN expression in 46 BC and 21 mammary fibroadenoma or fibroadenomatoid hyperplasia tissue samples. SCIN expression was ablated in MDA-MB-231 and T-47D cells using lentivirus-mediated small interfering RNA technology. Cell proliferation was tested using Celigo and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Cell apoptosis was analyzed by measuring Caspase 3/7 activity and annexin-V staining. The results of the present study demonstrated that SCIN expression was elevated in BC tissues compared with mammary fibroadenoma or fibroadenomatoid hyperplasia tissues. Specifically, higher SCIN expression was observed in Ki-67-positive BC tissues (78.6%) compared with Ki-67-negative BC tissues. Furthermore, knockdown of SCIN expression in the BC cell lines significantly suppressed cell proliferation and induced apoptosis. The data presented in the present study indicate that SCIN serves an important role in the development of breast cancer.
Collapse
Affiliation(s)
- Wenjing Jian
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China.,Department of Thyroid and Breast Surgery, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Xiaoli Zhang
- Central Laboratory, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Jiguo Wang
- Department of Medical Oncology, Baoan District Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518133, P.R. China
| | - Yunlong Liu
- Department of Thyroid and Breast Surgery, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Chuting Hu
- Department of Thyroid and Breast Surgery, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Xianming Wang
- Department of Thyroid and Breast Surgery, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Renbin Liu
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
25
|
Sun Z, Zheng L, Liu X, Xing W, Liu X. Sinomenine inhibits the growth of melanoma by enhancement of autophagy via PI3K/AKT/mTOR inhibition. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2413-2421. [PMID: 30122899 PMCID: PMC6084074 DOI: 10.2147/dddt.s155798] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Melanoma is a common skin tumor in adults with high metastasis and mortality rates. Thus, finding a better effective approach to treat melanoma has become very urgent. Sinomenine (SIN), the major active compound of Sinomenium acutum, has shown antitumorigenic activities in certain cancers. However, its role in melanoma remains unclear. Purpose This study aimed to explore the effects of SIN on melanoma in vitro and in vivo, in addition to exploring the underlying mechanism. Methods Mouse melanoma cell B16-F10 treated by SIN was analyzed by CCK8 assay and flow cytometry. Melanoma xenograft model was then established by subcutaneously injection with B16-F10 cells. Tumor growth was measured by immunohistochemistry. To further investigate the relative mechanism, the autophagy and PI3K/Akt/mTOR pathway were examined by immunofluorescence and Western blot. Results Our results revealed that SIN dose dependently inhibited the proliferation of B16-F10 cells in vitro and attenuated melanoma growth in vivo. In addition, SIN treatment promoted the apoptosis of B16-F10 cells in a dose-dependent manner, as demonstrated by the increase in apoptotic cells, Bax/Bcl-2 ratio, and caspase-3 activity. Moreover, preconditioning with SIN dramatically enhanced autophagy activity by increasing Beclin-1 and LC3II/LC3I expression, in addition to decreasing p62 expression and augmenting the number of LC3 puncta, in B16-F10 cells. More importantly, autophagy inhibitor chloroquine partly abolished SIN’s effects on cell growth and apoptosis. Furthermore, our results showed that SIN-triggered activation of autophagy was mediated by PI3K/Akt/mTOR signaling pathway. Conclusion Our study has identified a novel function of SIN and provided a molecular basis for potential applications of SIN in the treatment of melanoma and other cancers.
Collapse
Affiliation(s)
- Zheng Sun
- Department of Dermatology and Venereology, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lingling Zheng
- Department of Dermatology and Venereology, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xujun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Wenlong Xing
- Department of Cardiovasology, Beijing Chinese Medicine Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xinhai Liu
- Department of Plastic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China,
| |
Collapse
|