1
|
Huang HY, Lin LZY, Lu XY, Jiang Y. F-α-DDB-derivative, a novel synthetic of bifendate, plus epirubicin improves antitumor efficacy against triple negative breast cancer without additional cardiotoxicity. Discov Oncol 2025; 16:690. [PMID: 40338489 PMCID: PMC12061825 DOI: 10.1007/s12672-025-02545-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Triple negative breast cancer (TNBC) refers to a molecular subtype of breast cancers (BC) with high rate of distant metastases and poor prognosis. Epirubicin (EPI) is widely used for the therapy of TNBC but it's limited in clinical use due to cardiotoxicity and chemotherapy resistance. We previously identified F-α-DDB-derivative, a novel synthetic of bifendate, as a potential agent to improve the therapeutic efficacy of TNBC in combination with EPI since F-α-DDB-derivative inhibited MDA-MB-468, but not as much as EPI. In this study, we investigated the antitumor activity of F-α-DDB-derivative in combination with EPI against TNBC in vitro and in vivo and whether co-treatment would induce additional cardiotoxicity. In human TNBC MDA-MB-468 cells, application of F-α-DDB-derivative (11.5, 23.0, 46.0 μg/ml) in combination with EPI (1.5, 3.0, 6.0 μg/ml) exhibited great inhibition on cell viability and cell proliferation. Additionally, F-α-DDB-derivative (5.75, 11.5, 23.0, 46.0 μg/ml) interacted with EPI (0.75, 1.5, 3.0, 6.0 μg/ml) synergistically to induce apoptosis in MDA-MB-468 cells. This suggests that F-α-DDB-derivative may be more sensitive to apoptotic pathways. Furthermore, we revealed that co-administration of EPI and F-α-DDB-derivative (ip, once every other day for 14 days) significantly increased the therapeutic efficacy of EPI (2.0 mg/kg) or F-α-DDB-derivative (20.0 mg/kg) in mice harboring MDA-MB-468 cell xenografts without additional cardiotoxicity compared to that in EPI monotherapy group. These results implicate that co-treatment of EPI and F-α-DDB-derivative may be a potential therapeutic approach for the treatment of TNBC.
Collapse
Affiliation(s)
- Hai-Yi Huang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lisa Zong Yong Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin-Yi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China.
| |
Collapse
|
2
|
Pei S, Zhang D, Li Z, Liu J, Li Z, Chen J, Xie Z. The Role of the Fox Gene in Breast Cancer Progression. Int J Mol Sci 2025; 26:1415. [PMID: 40003882 PMCID: PMC11855465 DOI: 10.3390/ijms26041415] [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: 11/26/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Forkhead box (FOX) genes are a family of transcription factors that participate in many biological activities, from early embryogenesis to the formation of organs, and from regulation of glucose metabolism to regulation of longevity. Given the extensive influence in the multicellular process, FOX family proteins are responsible for the progression of many types of cancers, especially lung cancer, breast cancer, prostate cancer, and other cancers. Breast cancer is the most common cancer among women, and 2.3 million women were diagnosed in 2020. So, various drugs targeting the FOX signaling pathway have been developed to inhibit breast cancer progression. While the role of the FOX family gene in cancer development has not received enough attention, discovering more potential drugs targeting the FOX signaling pathway is urgently demanded. Here, we review the main members in the FOX gene family and summarize their signaling pathway, including the regulation of the FOX genes and their effects on breast cancer progression. We hope this review will emphasize the understanding of the role of the FOX gene in breast cancer and inspire the discovery of effective anti-breast cancer medicines targeting the FOX gene in the future.
Collapse
Affiliation(s)
- Shaoxuan Pei
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Dechun Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Ziyi Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
| |
Collapse
|
3
|
Gao C, Li X, Liu T, Wang W, Wu J. An overview of phenylsulfonylfuroxan-based nitric oxide donors for cancer treatment. Bioorg Chem 2025; 154:108020. [PMID: 39657549 DOI: 10.1016/j.bioorg.2024.108020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
Nitric oxide (NO) is a gaseous molecule integral to numerous physiological processes, including tumor modulation, cardiovascular regulation, and systemic physiological functions. Its dual role in promoting and inhibiting tumor growth makes it a focal point of contemporary oncological research. Phenylsulfonylfuroxan, a classical NO donor, has been shown to significantly elevate NO levels, thereby inducing apoptosis and inhibiting proliferation and metastasis in tumor cells. It enhances the efficacy of chemotherapy, radiotherapy, and immunotherapy, reverses multidrug resistance (MDR), and impedes tumor progression. Notably, phenylsulfonylfuroxan have the ability to trigger ferroptosis in cancer cells by binding covalently to inhibit glutathione peroxidase 4 (GPX4). Recent developments in phenylsulfonylfuroxan-based therapies have positioned them as crucial in the advancement of cancer treatment modalities. This review elucidates the mechanism by which phenylsulfonylfuroxan releases NO and summarizes the significant advancements over the past 16 years in the research and development of phenylsulfonylfuroxan conjugates with various anticancer agents for targeted cancer therapy.
Collapse
Affiliation(s)
- Chao Gao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Xingyu Li
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Tong Liu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Wanning Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
4
|
Luo Z, Yin F, Wang X, Kong L. Progress in approved drugs from natural product resources. Chin J Nat Med 2024; 22:195-211. [PMID: 38553188 DOI: 10.1016/s1875-5364(24)60582-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 04/02/2024]
Abstract
Natural products (NPs) have consistently played a pivotal role in pharmaceutical research, exerting profound impacts on the treatment of human diseases. A significant proportion of approved molecular entity drugs are either directly derived from NPs or indirectly through modifications of NPs. This review presents an overview of NP drugs recently approved in China, the United States, and other countries, spanning various disease categories, including cancers, cardiovascular and cerebrovascular diseases, central nervous system disorders, and infectious diseases. The article provides a succinct introduction to the origin, activity, development process, approval details, and mechanism of action of these NP drugs.
Collapse
Affiliation(s)
- Zhongwen Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Fucheng Yin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaobing Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
5
|
Wang J, Liu Y, Guo Y, Liu C, Yang Y, Fan X, Yang H, Liu Y, Ma T. Function and inhibition of P38 MAP kinase signaling: Targeting multiple inflammation diseases. Biochem Pharmacol 2024; 220:115973. [PMID: 38103797 DOI: 10.1016/j.bcp.2023.115973] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Inflammation is a natural host defense mechanism that protects the body from pathogenic microorganisms. A growing body of research suggests that inflammation is a key factor in triggering other diseases (lung injury, rheumatoid arthritis, etc.). However, there is no consensus on the complex mechanism of inflammatory response, which may include enzyme activation, mediator release, and tissue repair. In recent years, p38 MAPK, a member of the MAPKs family, has attracted much attention as a central target for the treatment of inflammatory diseases. However, many p38 MAPK inhibitors attempting to obtain marketing approval have failed at the clinical trial stage due to selectivity and/or toxicity issues. In this paper, we discuss the mechanism of p38 MAPK in regulating inflammatory response and its key role in major inflammatory diseases and summarize the synthetic or natural products targeting p38 MAPK to improve the inflammatory response in the last five years, which will provide ideas for the development of novel clinical anti-inflammatory drugs based on p38 MAPK inhibitors.
Collapse
Affiliation(s)
- Jiahui Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yongjian Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yushi Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Cen Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuping Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoxiao Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongliu Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Tao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
6
|
Ampawong S, Tirawanchai N, Kanjanapruthipong T, Fongsodsri K, Tuentam K, Isarangkul D, Aramwit P. Sericin enhances ammonia detoxification by promotes urea cycle enzyme genes and activates hepatic autophagy in relation to CARD-9/MAPK pathway. Heliyon 2023; 9:e21563. [PMID: 38027599 PMCID: PMC10654145 DOI: 10.1016/j.heliyon.2023.e21563] [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: 03/23/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Urea cycle is an important metabolic process that initiates in liver mitochondria and converts ammonia to urea. The impairment of ammonia detoxification, both primary and secondary causes, lead to hyperammonemia, a life-threatening condition affecting to the brain. Current treatments are not enough effective. In addition, our recent proteomics study in hypercholesterolemic rat model demonstrated that sericin enhances hepatic nitrogenous waste removal through carbamoyl-phosphate synthase 1 (CPS-1), aldehyde dehydrogenase-2 (ALDH-2), and uricase proteins. However, the underlining mechanisms regard to this property is not clarified yet. Therefore, the present study aims to examine the effect of sericin on urea cycle enzyme genes (CPS-1 and ornithine transcarbamylase; OTC) and proteins (mitogen-activated protein kinase; MAPK, caspase recruitment domain-containing protein 9; CARD-9, Microtubule-associated protein light chain 3; LC-3), which relate to urea production and liver homeostasis in hepatic cell line (HepG2) and hypercholesterolemic rat treated with or without sericin. qRT-PCR, immunohistochemistry, and electron microscopy techniques were performed. In vitro study determined that high dose of sericin at 1 mg/ml increased liver detoxification enzyme (Cytochrome P450 1A2; CYP1A2 and ALDH-2) and urea cycle enzyme (CPS-1 and OTC) genes. Both in HepG2 cell and rat liver mitochondria, sericin significantly downregulated CARD-9 (apoptotic protein) expression while upregulated MAPK (hepatic homeostasis protein) and LC-3 (autophagic protein) expressions. Hence, it might be concluded that sericin promotes ammonia detoxification by both increases urea cycle enzyme genes and enhances hepatic autophagy in associated with CARD-9/MAPK pathway (as shown by their own negative relationship). This study presents another beneficial property of sericin to develop an upcoming candidate for ammonia toxicity alleviation and liver function improvement.
Collapse
Affiliation(s)
- Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Napatara Tirawanchai
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Road, Bangkoknoi, Bangkok, 10700, Thailand
| | - Tapanee Kanjanapruthipong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Kamonpan Fongsodsri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Khwanchanok Tuentam
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Duangnate Isarangkul
- Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pornanong Aramwit
- Center of Excellence in Bioactive Resources for Innovative Clinical Applications and Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, PhayaThai Road, Phatumwan, Bangkok, 10330, Thailand
- The Academy Science, The Royal Society of Thailand, Dusit, Bangkok, 10330, Thailand
| |
Collapse
|
7
|
Wang F, Liao R, Wang X, Xiong G, Zhang B, Li J, Wu D, Chen Y, Zhou X, Gu X, Qi Q, Li C. N-3, a novel synthetic derivative of bifendate, inhibits metastasis of triple-negative breast cancer via decreasing p38-regulated FOXC1 protein stability. Biochem Pharmacol 2023; 215:115729. [PMID: 37558004 DOI: 10.1016/j.bcp.2023.115729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/16/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with high invasiveness, metastatic potential, and poor prognosis. Epithelial-mesenchymal transition (EMT) is pivotal in TNBC progression, becoming a promising target for TNBC treatment. Our study evaluated N-3, a novel synthetic bifendate derivative, which inhibited the EMT-associated migration and invasion of MDA-MB-231 and 4T1 TNBC cells. The results were consistent with the suppression of FOXC1 expression and transcriptional activity. Additional studies indicated that N-3 reduced the protein stability of FOXC1 by enhancing ubiquitination and degradation. Moreover, N-3 downregulated p-p38 expression and FOXC1 interaction, decreasing the stability of p38-regulated FOXC1. Further, N-3 blocked TNBC metastasis with an artificial lung metastasis model in vivo, related to FOXC1 suppression and EMT. These results highlight the potential of N-3 as a TNBC metastasis treatment. Therefore, FOXC1 regulation could be a novel targeted therapeutic strategy for TNBC metastasis.
Collapse
Affiliation(s)
- Fan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Rong Liao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xin Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Pharmacy, Xuzhou City Hospital of TCM, Xuzhou 221010, Jiangsu, China
| | - Guixiang Xiong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Beibei Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Juan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Dengpan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yan Chen
- Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 550025, China
| | - Xueyan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Qi Qi
- MOE Key Laboratory of Tumor Molecular Biology, Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
8
|
Network Pharmacology/Metabolomics-Based Validation of AMPK and PI3K/AKT Signaling Pathway as a Central Role of Shengqi Fuzheng Injection Regulation of Mitochondrial Dysfunction in Cancer-Related Fatigue. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5556212. [PMID: 34326918 PMCID: PMC8302405 DOI: 10.1155/2021/5556212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/06/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022]
Abstract
Chinese herbal medicines have multiple targets and properties, and their use in multidisciplinary cancer therapies has consequently received increasing attention. Here, we have investigated the possible active ingredients associated with cancer-related fatigue (CRF) in the Shengqi Fuzheng Injection (SFI). In vitro cell models were used to measure the regulation effects of SFI on CRF. Metabolomic analysis was used to identify the potential genes and pathways in C2C12 mouse myoblasts treated with SFI, and the interaction of compounds and CRF targets was predicted using network pharmacology and molecular docking analyses. The putative pathways were further verified using immuno-blotting assays. The results showed that SFI significantly inhibited muscle cell apoptosis and increased the mitochondrial membrane potential of muscle cells. The network pharmacology analysis results identified 36 candidate compounds, and 244 potential targets were yielded by SFI, and they shared 10 key targets associated with cancer-related fatigue. According to the enrichment analysis and experimental validation, SFI might ameliorate muscle cell mitochondrial function by activating AMPK and inhibiting the PI3K/Akt signaling pathways, and the expression changes of mitochondrial metabolic enzymes MnSOD and apoptosis-associated proteins Bax and Bcl-2 were also triggered. The functions and mechanisms of SFI in anticancer-related fatigue were found here to be at least partly due to the targeting of the AMPK and PI3K/Akt signaling pathways, and this has highlighted new potential applications for network pharmacology when researching Chinese Medicines.
Collapse
|
9
|
Song N, Ma J, Hu W, Guo Y, Hui L, Aamer M, Ma J. Lappaconitine hydrochloride inhibits proliferation and induces apoptosis in human colon cancer HCT-116 cells via mitochondrial and MAPK pathway. Acta Histochem 2021; 123:151736. [PMID: 34058516 DOI: 10.1016/j.acthis.2021.151736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Lappaconitine hydrochloride (LH), as a new synthetic alkaloid, exhibits antitumor activity, whereas its antitumor effect on colorectal cancer (CRC) has not been investigated. In this study, the effect of LH on HCT-116 cell proliferation and apoptosis in vivo and in vitro and underlying molecular mechanism were explored. The Cell Counting Kit-8 (CCK-8) was used to assess cell viability. Morphological change was observed by Hoechst 33342 staining. Cell cycle and apoptosis were performed using a flow cytometer. The western blot method was used to screen for related protein expression. The mitochondrial membrane potential (MMP) was confirmed using the 5, 5, 6, 6'-tetrachloro-1, 1', 3, 3'-tetraethylbenzimi-dazolyl carbo cyanine iodide (JC-1) staining assay. Reactive oxygen species (ROS) was evaluated by a 20-70-dichlorofluorescein diacetate (DCFH-DA) staining assay. The antitumor effect was evaluated in vivo by the xenograft HCT-116 model. The results showed that LH significantly inhibited cell viability in a time- and concentration-dependent manner. LH induced apoptosis and S phase cell cycle arrest. LH promoted the reduction of MMP and ROS accumulation. Moreover, LH activated the mitochondrial and MAPK pathway. The experiments in vivo showed that LH had significant antitumor effect in tumor-bearing mice, and had virtually no effect on the weight and internal organs of the mice. In conclusion, LH could induce apoptosis in HCT-116 cells through mitochondrial and MAPK signaling pathways. LH may be a promising treatment for CRC.
Collapse
Affiliation(s)
- Na Song
- College of Life Science, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Junyi Ma
- College of Life Science, Northwest Normal University, Lanzhou, Gansu, 730070, China.
| | - Wei Hu
- College of Life Science, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Yongyue Guo
- College of Life Science, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Ling Hui
- Gansu Province Center of Medical Genetics, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, Gansu, 730070, China.
| | - Mohamed Aamer
- College of Life Science, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Jun Ma
- Key Laboratory of Stem Cells and Gene Drug of Gansu Provincial, The 940(th) Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, 730070, China
| |
Collapse
|
10
|
Lin Z, Miao D, Xu Q, Wang X, Yu F. A novel focal adhesion related gene signature for prognostic prediction in hepatocellular carcinoma. Aging (Albany NY) 2021; 13:10724-10748. [PMID: 33850056 PMCID: PMC8064231 DOI: 10.18632/aging.202871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous disease. Reduced expression of focal adhesion is considered as an important prerequisite for tumor cell invasion and metastasis. However, the prognostic value of focal adhesion related genes in HCC remains to be further determined. In this study, RNA expression profiles were downloaded from public databases. A five focal adhesion related gene signature model was established by the least absolute shrinkage and selection operator Cox regression analysis, which categorized patients into high- and low-risk groups. Multivariate Cox regression analysis showed that the risk score was an independent predictor for overall survival. Single-sample gene set enrichment analysis revealed that immune status was different between the two risk groups, and tumor-related pathways were enriched in high-risk group. The risk score was significantly associated with tumor grade, tumor stage, immune scores, and immune infiltrate types. Pearson correlation showed that the expression level of prognostic genes was associated with anti-tumor drug sensitivity. Besides, the mRNA and protein expression of prognostic genes was significantly different between HCC tissues and adjacent non-tumorous tissues in our separate cohort. Taken together, a novel focal adhesion related gene signature can be used for prognostic prediction in HCC, which may be a therapeutic alternative.
Collapse
Affiliation(s)
- Zhuo Lin
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, Zhejiang, China
| | - Dan Miao
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qian Xu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Wang
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou, Zhejiang, China
| | - Fujun Yu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|