1
|
Liu Y, Wu Z, jin X, Ji M, Huang T, Meng P, Xu T, You W, Zhao Y, Ye F, Wu X. Paeonol Improves Cardiac Remodelling in MI Mice by Suppressing NOX2 mRNA Expression to Mitigate Oxidative Stress and Mitochondrial Dysfunction. J Cell Mol Med 2025; 29:e70563. [PMID: 40344316 PMCID: PMC12061634 DOI: 10.1111/jcmm.70563] [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: 12/01/2024] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Myocardial infarction (MI), a primary contributor to mortality from cardiovascular diseases, continues to pose a significant challenge in clinical treatment. In this study, our objective was to investigate the cardioprotective effects of paeonol (PAE) on mice with MI, and to delve into the precise mechanisms underlying these effects. We developed the MI model by ligating the left anterior descending artery in mice and replicated this model in vitro by stimulating H9C2 cells with levarterenol (LN). Cardiac function, infarct size, cardiomyocyte size, apoptosis, and mitochondrial structure were evaluated through echocardiography, Masson's trichrome staining, WGA staining, TUNEL assay, and electron microscopy, respectively. Colorimetry, Western blotting, flow cytometry, RT-PCR, and the dual-luciferase reporter assay were employed to explore the underlying mechanisms. Compared with the model group, PAE significantly ameliorated cardiac dysfunction and hypertrophy, diminished infarct size, cardiomyocyte hypertrophy, and apoptosis, mitigated mitochondrial structural damage, lowered levels of malondialdehyde and NOX2, reduced ROS production, and NOX activity, while enhancing the activities of T-SOD, GSH-PX, and mitochondrial complexes I-V in mice with MI or H9C2 cells subjected to LN intervention. Ultimately, PAE was found to negatively regulate the transcription of NOX2 mRNA in H9C2 cells, partly through inhibition of phospho-STAT3-Y705 protein expression. These results imply that PAE's transcriptional inhibition of NOX2 mRNA expression primarily confers a cardioprotective effect, mitigating myocardial remodelling following MI by improving oxidative stress and mitochondrial dysfunction. This indicates that PAE holds therapeutic promise for the treatment of patients post-MI.
Collapse
Affiliation(s)
- Yun Liu
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
- Department of CardiologyThe Eighty‐First Hospital of PLA Affiliated With Anhui Medical UniversityNanjingChina
| | - Zhiming Wu
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Xiaoping jin
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Meili Ji
- Department of GeriatricNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Tianyi Huang
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Peina Meng
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Tian Xu
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Wei You
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Yanfang Zhao
- Department of CardiologyThe Eighty‐First Hospital of PLA Affiliated With Anhui Medical UniversityNanjingChina
| | - Fei Ye
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| | - Xiangqi Wu
- Department of CardiologyNanjing First Hospital, Nanjing Medical UniversityNanjingChina
| |
Collapse
|
2
|
Zhang S, Xie X, Zhao J, Jiang Y, Huang C, Li Q, Xia B, Yin L, Yuan X, You Q. Andrographolide and its Derivatives in Cardiovascular Disease: A Comprehensive Review. PLANTA MEDICA 2025; 91:259-270. [PMID: 40054492 DOI: 10.1055/a-2542-0756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Cardiovascular disease is one of the main causes of mortality worldwide. Andrographolide represents an important category of natural phytochemicals that has significant therapeutic potential in various conditions such as acute lung injury, heart disease, and viral infections due to its anti-oxidative, anti-inflammatory, and anti-apoptotic properties. This compound plays a protective role in human pathophysiology. This review provides a comprehensive overview of the effects of andrographolide on cardiovascular disease and examines its essential roles and mechanisms in cardiovascular disease and other vascular dysfunctions. The data collected in this review serve as a comprehensive reference for the role of andrographolide in cardiovascular disease and provide valuable insights for further research and the development of andrographolide as a novel therapeutic approach for cardiovascular disease.
Collapse
Affiliation(s)
- Shenjie Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaokai Xie
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Juan Zhao
- Department of Cardiology, Tongzhou People's Hospital, Nantong, China
| | - Yilong Jiang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Le Yin
- Department of Cardiology, Tongzhou People's Hospital, Nantong, China
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
3
|
Tang K, Deng W, Huang Z, Chen S, Zhu Z, Lin S, Zhong L, Zheng Q, Zhao W, Zhang Z, Mo L. Neoandrographolide inhibits mature osteoclast differentiation to alleviate bone loss and treat osteoporosis. Front Pharmacol 2025; 16:1466057. [PMID: 40008134 PMCID: PMC11851123 DOI: 10.3389/fphar.2025.1466057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/02/2025] [Indexed: 02/27/2025] Open
Abstract
Background Osteoporosis (OP), as the prevalent systemic metabolic bone disease worldwide, progresses insidiously and slowly. The clinical discomfort and complications associated with OP impose a significant burden on patients. Therefore, finding more effective treatments for OP remains an urgent challenge. Method We first conducted in vitro experiments to determine whether Neoandrographolide (NEO) exhibits cytotoxic or proliferative effects on bone marrow macrophages (BMMs) and to explore the specific timeframe during which NEO exerts its inhibitory action on osteoclast (OC) differentiation. Through Reverse Transcription Polymerase Chain Reaction (RT-PCR) and Western blot analysis, we examined the relative expression levels of genes and proteins associated with OC differentiation like CTSK,c-Fos,MMP9,NFATc1, and verified the underlying mechanisms. Finally, we performed in vivo experiments to further investigate the inflammation. Results NEO exhibits no significant cytotoxic effects on BMMs at concentrations less than or equal to 30 μM while exerting inhibitory effects on OC differentiation during its early and middle stages. RT-PCR and Western blot results reveal that NEO suppresses the expression of genes and proteins including CTSK,c-Fos,MMP9,NFATc1. Western blot findings also indicate that NEO inhibits the phosphorylation of ERK, P38, JNK, and P65 but does not reverse the degradation of IκB-α. Additionally, NEO affects the phosphorylation of proteins in the PI3K/AKT, GSK3β, and PPARγ signaling pathways, demonstrating that NEO can inhibit OC formation through multiple pathways and targets. In vivo experiments further validated the in vitro findings by constructing an OP model, showing that NEO can mitigate bone loss induced by OC differentiation. Conclusion NEO has the potential to serve as a therapeutic agent for OP by targeting multiple sites and inhibiting the formation of mature OC through various signaling pathways.
Collapse
Affiliation(s)
- Kai Tang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Deng
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiying Huang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Eighth School of Clinical Medicine of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Simin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zilin Zhu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shukun Lin
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lubin Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Quanxin Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenhua Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhida Zhang
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Mo
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Research Institute for Orthopedics and Traumatology of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Lu H, Zhang J, Wang Y. Identification of the Pharmacological Components and Its Targets of Sanghuang by Integration of Nontarget Metabolomics and Network Pharmacology Analysis. Biomed Chromatogr 2025; 39:e6066. [PMID: 39748251 DOI: 10.1002/bmc.6066] [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: 07/31/2024] [Revised: 10/09/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025]
Abstract
The objective of this study is to comprehensively to identify the core pharmacological components and their respective targets of three medicinal fungi Sanghuangs including Sanghuangporus vaninii (SV), Sanghuangporus lonicericola (SL), and Inonotus hispidus (IH). Metabolomics analysis indicated that a total of 495 and 660 differential metabolites were obtained in mycelium and fermentation broth samples among three Sanghuangs, respectively. The network pharmacology analysis showed that 6-[1]-ladderane hexanol, R-nostrenol, candidone, ellagic acid, and quercetin were the overlapping active ingredients of three Sanghuang species for diabetes mellitus, immune system disease, and neoplasm. Certonardosterol A, dalamid, and ethylene brassylate are unique active ingredients in SV, and certonardosterol K, kaempferide, and esculetin are unique active ingredients in SL. Asbestinine, neoandrographolide, isosakuranetin, and daucosterin are unique active ingredients in IH. Accordingly, the common core targets of active ingredients of the three Sanghuangs were ESR1, PIK3CA, and LYN. PRKCA, EGFR, and STAT3 were the unique targets of SV, SL, and IH, respectively. The primary active components and their respective targets, in addition to the component-target interaction of Sanghuangs that have been identified in the present study, provide a foundation for future research on the prevention and treatment of disease using Sanghuangs.
Collapse
Affiliation(s)
- Hengqian Lu
- School of Life Sciences, Anhui University, Hefei, Anhui, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
- Wuhu Dongyuan New Rural Co. Ltd., Wuhu, China
| | - Jintao Zhang
- School of Life Sciences, Anhui University, Hefei, Anhui, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, Anhui, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui, China
- Anhui Province Joint Construction Discipline key Laboratory of Nanobody Technology, Hefei, China
| |
Collapse
|
5
|
Yu K, Liang P, Yu H, Liu H, Guo J, Yan X, Li Z, Li G, Wang Y, Wang C. Integrating Transcriptome and Chemical Analyses to Provide Insights into Biosynthesis of Terpenoids and Flavonoids in the Medicinal Industrial Crop Andrographis paniculate and Its Antiviral Medicinal Parts. Molecules 2024; 29:852. [PMID: 38398604 PMCID: PMC10893308 DOI: 10.3390/molecules29040852] [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: 01/14/2024] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Andrographis paniculata is a medicinal plant traditionally used to produce diterpene lactones and flavonoids, which possess various biological activities. Widely distributed in China, India, and other Southeast Asia countries, A. paniculata has become an important economic crop, significantly treating SARS-CoV-2, and is being cultivated on a large scale in southern China. The biosynthesis of active ingredients in A. paniculata are regulated and controlled by genes, but their specific roles are still not fully understood. To further explore the growth regulation factors and utilization of its medicinal parts of this industrial crop, chemical and transcriptome analyses were conducted on the roots, stems, and leaves of A. paniculata to identify the biosynthesis pathways and related candidate genes of the active ingredients. The chemical analysis revealed that the main components of A. paniculata were diterpene lactones and flavonoids, which displayed potential ability to treat SARS-CoV-2 through molecular docking. Moreover, the transcriptome sequencing annotated a total of 40,850 unigenes, including 7962 differentially expressed genes. Among these, 120 genes were involved in diterpene lactone biosynthesis and 60 genes were involved in flavonoid biosynthesis. The expression of diterpene lactone-related genes was the highest in leaves and the lowest in roots, consistent with our content determination results. It is speculated that these highly expressed genes in leaves may be involved in the biosynthesis pathway of diterpenes. Furthermore, two class Ⅰ terpene synthases in A. paniculata transcriptome were also annotated, providing reference for the downstream pathway of the diterpene lactone biosynthesis. With their excellent market value, our experiments will promote the study of the biosynthetic genes for active ingredients in A. paniculata and provide insights for subsequent in vitro biosynthesis.
Collapse
Affiliation(s)
- Kuo Yu
- School of Medicine, Foshan University, Foshan 528225, China; (K.Y.); (P.L.); (H.L.); (J.G.); (G.L.)
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (H.Y.); (X.Y.); (Z.L.)
| | - Pengjie Liang
- School of Medicine, Foshan University, Foshan 528225, China; (K.Y.); (P.L.); (H.L.); (J.G.); (G.L.)
| | - Heshui Yu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (H.Y.); (X.Y.); (Z.L.)
| | - Hui Liu
- School of Medicine, Foshan University, Foshan 528225, China; (K.Y.); (P.L.); (H.L.); (J.G.); (G.L.)
| | - Jialiang Guo
- School of Medicine, Foshan University, Foshan 528225, China; (K.Y.); (P.L.); (H.L.); (J.G.); (G.L.)
| | - Xiaohui Yan
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (H.Y.); (X.Y.); (Z.L.)
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (H.Y.); (X.Y.); (Z.L.)
| | - Guoqiang Li
- School of Medicine, Foshan University, Foshan 528225, China; (K.Y.); (P.L.); (H.L.); (J.G.); (G.L.)
| | - Ying Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Chunhua Wang
- School of Medicine, Foshan University, Foshan 528225, China; (K.Y.); (P.L.); (H.L.); (J.G.); (G.L.)
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (H.Y.); (X.Y.); (Z.L.)
| |
Collapse
|
6
|
Casper E. The crosstalk between Nrf2 and NF-κB pathways in coronary artery disease: Can it be regulated by SIRT6? Life Sci 2023; 330:122007. [PMID: 37544377 DOI: 10.1016/j.lfs.2023.122007] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 06/26/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Oxidative stress and inflammation are major mechanisms responsible for the progression of CAD. Nuclear transcription factor erythroid-2 related factor 2 (Nrf2) is a transcription factor that modulates the cellular redox status. Nrf2 upregulation increases the expression of antioxidant genes, decreases the expression of Nuclear factor-kappa B (NF-kB), and increases free radical metabolism. Activated NF-kB increases the production of inflammatory cytokines causing endothelial dysfunction. The two pathways of Nrf2 and NF-kB can regulate the expression of each other. Foremost, the Nrf2 pathway can decrease the level of active NF-κB by increasing the level of antioxidants and cytoprotective enzymes. Furthermore, the Nrf2 pathway prevents IκB-α degradation, an inhibitor of NF-kB, and thus inhibits NF-κB mediated transcription. Also, NF-kB transcription inhibits Nrf2 activation by reducing the antioxidant response element (ARE) transcription. Sirtuin 6 (SIRT6) is a member of the Sirtuins family that was found to protect against cardiovascular diseases. SIRT6 can suppress the production of Reactive oxygen species (ROS) through deacetylation of NRF2 which results in NRF2 activation. Furthermore, SIRT6 can inhibit the inflammatory process through the downregulation of NF-kB transcription. Therefore, targeting sirtuins could be a therapeutic strategy to treat CAD. This review describes the potential role of SIRT6 in regulating the crosstalk between NRF2 and NF-kB signaling pathways in CAD.
Collapse
Affiliation(s)
- Eman Casper
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
7
|
Lai CH, Van Thao D, Tsai BCK, Hsieh DJY, Chen MYC, Kuo WW, Kuo CH, Lu SY, Liao SC, Lin KH, Huang CY. Insulin-like growth factor II receptor alpha overexpression in heart aggravates hyperglycemia-induced cardiac inflammation and myocardial necrosis. ENVIRONMENTAL TOXICOLOGY 2023; 38:676-684. [PMID: 36462176 DOI: 10.1002/tox.23717] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/07/2022] [Accepted: 11/20/2022] [Indexed: 06/17/2023]
Abstract
Diabetes-induced cardiovascular complications are mainly associated with high morbidity and mortality in patients with diabetes. Insulin-like growth factor II receptor α (IGF-IIRα) is a cardiac risk factor. In this study, we hypothesized IGF-IIRα could also deteriorate diabetic heart injury. The results presented that both in vivo transgenic Sprague-Dawley rat model with specific IGF-IIRα overexpression in the heart and in vitro myocardium H9c2 cells were used to investigate the negative function of IGF-IIRα in diabetic hearts. The results showed that IGF-IIRα overexpression aided hyperglycemia in creating more myocardial injury. Pro-inflammatory factors, such as Tumor necrosis factor-alpha, Interleukin-6, Cyclooxygenase-2, Inducible nitric oxide synthase, and Nuclear factor-kappaB inflammatory cascade, are enhanced in the diabetic myocardium with cardiac-specific IGF-IIRα overexpression. Correspondingly, IGF-IIRα overexpression in the diabetic myocardium also reduced the PI3K-AKT survival axis and activated mitochondrial-dependent apoptosis. Finally, both ejection fraction and fractional shortening were be significantly decrease in diabetic rats with cardiac-specific IGF-IIRα overexpression. Overall, all results provid clear evidence that IGF-IIRα can enhance cardiac damage and is a harmful factor to the heart under high-blood glucose conditions. However, the pathophysiology of IGF-IIRα under different stresses and its downstream regulation in the heart still require further research.
Collapse
Affiliation(s)
- Chin-Hu Lai
- Division of Cardiovascular Surgery, Department of Surgery, Taichung Armed Force General Hospital, Taichung, Taiwan
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
- Center of General Education is division, National Defense Medical Center, Taipei, Taiwan
| | - Dao Van Thao
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Michael Yu-Chih Chen
- Department of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, Virginia, USA
| | - Shang-Yeh Lu
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Chieh Liao
- Department of Social Medicine, School of Medicine, China Medical University, Taichung, Taiwan
| | - Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung, Taiwan
- Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| |
Collapse
|
8
|
Qiu J, Zhang Z, Hu A, Zhao P, Wei X, Song H, Yang J, Li Y. Integrating UPLC-HR-MS/MS, Network Pharmacology, and Experimental Validation to Uncover the Mechanisms of Jin'gan Capsules against Breast Cancer. ACS OMEGA 2022; 7:28003-28015. [PMID: 35990498 PMCID: PMC9386888 DOI: 10.1021/acsomega.2c01921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
In the theory of traditional Chinese medicine (TCM), "liver-qi" stagnation and heat-induced toxicity represent the main etiologies of breast cancer. Recently, several TCMs with heat-clearing and detoxification efficacy have shown inhibitory effects on breast cancer. Jin'gan capsules (JGCs), initially approved to treat colds in China, are a heat-clearing and detoxification TCM formula. However, the anticancer activity of JGCs against breast cancer and its underlying mechanisms remain unclear. First, we assessed the antiproliferative activity of JGCs in breast cancer cell lines and evaluated their effects on cell apoptosis and the cell cycle by flow cytometry. Furthermore, we identified the potential bioactive components of JGCs and their corresponding target genes and constructed a bioactive compound-target interaction network by ultra-performance liquid chromatography-high-resolution tandem mass spectrometry (UPLC-HR-MS/MS) and network pharmacology analysis. Finally, the underlying mechanism was investigated through gene function enrichment analysis and experimental validation. We found that JGCs significantly inhibited breast cancer cell growth with IC50 values of 0.56 ± 0.03, 0.16 ± 0.03, and 0.94 ± 0.09 mg/mL for MDA-MB-231, MDA-MB-468, and MCF-7, respectively. In addition, JGC treatment dramatically induced apoptosis and S phase cell cycle arrest in breast cancer cells. Western blot analysis confirmed that JGCs could regulate the protein levels of apoptosis- and cell cycle-related genes. Utilizing UPLC-HR-MS/MS analysis and network pharmacology, we identified 7 potential bioactive ingredients in JGCs and 116 antibreast cancer targets. Functional enrichment analysis indicated that the antitumor effects of JGCs were strongly associated with apoptosis and the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway. Western blot analysis validated that JGC treatment markedly decreased the expression levels of p-JAK2, p-STAT3, and STAT3. Our findings suggest that JGCs suppress breast cancer cell proliferation and induce cell cycle arrest and apoptosis partly by inhibiting the JAK2/STAT3 signaling pathway, highlighting JGCs as a potential therapeutic candidate against breast cancer.
Collapse
Affiliation(s)
- Jianfei Qiu
- State
Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- State
Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| | - Zhiyin Zhang
- Guiyang
Hospital of Guizhou Aviation Industry Group, Guiyang 550025, China
| | - Anling Hu
- State
Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- State
Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| | - Peng Zhao
- State
Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- State
Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| | - Xuenai Wei
- State
Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- State
Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| | - Hui Song
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| | - Jue Yang
- State
Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- State
Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| | - Yanmei Li
- State
Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- State
Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- The
Key Laboratory of Chemistry for Natural Products of Guizhou Province
and Chinese Academic of Sciences & Key Laboratory of Endemic and
Ethnic Diseases, Ministry of Education & Key Laboratory of Medical
Molecular Biology of Guizhou Province, Guizhou
Medical University, Guiyang 550004, China
| |
Collapse
|