1
|
Zhu JF, Wang YQ, Yang SM, Wang YL, Hu Y, Dai XY. Exploring the mechanism of Bruceine D against cervical cancer by network pharmacology and the effect of Bruceine D on the EGFR pathway. J Pharm Biomed Anal 2025; 262:116887. [PMID: 40239560 DOI: 10.1016/j.jpba.2025.116887] [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: 01/10/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Cervical cancer (CC) remains a formidable challenge in oncology due to its high incidence and mortality rates. Despite recent advances in treatment, an immediate necessity exists for innovating advanced pharmacological interventions boasting augmented effectiveness. Bruceine D (BD), a quassinoid derived from the traditional Chinese medicinal plant Brucea javanica, has been demonstrated to possess notable anticancer properties against a range of malignant conditions, including lung, liver, leukemia, and pancreatic cancers. However, its specific effects on CC have not been thoroughly explored. This study sought to decode the effects of BD on CC through a combined method involving molecular docking analysis, network pharmacology, and data mining. From the PharmMapper database, we identified 58 potential targets of BD, and through GeneCards, we pinpointed 14 intersecting targets relevant to CC. A protein-protein interaction (PPI) network highlighted pivotal targets such as ESR1, HSP90AA1, ANXA5, EGFR, CASP7, and CCNA2. GO and KEGG enrichment analyses underscored significant biological processes and pathways, notably the EGFR signaling pathway. Molecular docking analysis revealed a strong binding affinity of BD to EGFR. Cell-based assays demonstrated that BD potently curtailed the viability, colony formation, adhesion, and mobility of Hela and Caski cells, escalating apoptosis in a dose-proportional manner. Supplementary evidence via western blot evaluations underscored BD's capability to obstruct the EGFR signaling pathway. These findings suggest that BD exerts potent anticancer effects against CC through multiple mechanisms, positioning it as a promising therapeutic agent for further investigation and clinical validation.
Collapse
Affiliation(s)
- Ju-Fan Zhu
- Institute of Organoid Technology, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yuan-Qiu Wang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Si-Meng Yang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yu-Li Wang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yan Hu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xin-Yue Dai
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
2
|
Shan L, Gong M, Zhai D, Meng X, Liu J, Lv X. Research progress of CD73-adenosine signaling regulating hepatocellular carcinoma through tumor microenvironment. J Exp Clin Cancer Res 2025; 44:161. [PMID: 40420185 PMCID: PMC12105175 DOI: 10.1186/s13046-025-03416-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025] Open
Abstract
Adenosine signaling pathway is a kind of signal regulation hub widely existing in human body, which is involved in a series of physiological processes such as energy supply of body cells. CD73 is a highly concerned signaling protein in purine adenosine pathway, and its role in tumor development and prognosis has been paid more and more attention in recent years, especially in hepatocellular carcinoma (HCC). In this paper, the specific mechanism by which CD73-adenosine signaling regulates tumor microenvironment (TME) of liver cancer tumors was analyzed in detail, highlighting the importance of this pathway as a therapeutic target to combat tumor immunosuppression and enhance the anti-tumor immune response to prevent and treat hepatocellular carcinoma (HCC). In addition, a variety of current targeted therapeutic strategies for adenosine metabolic pathways are summarized, including the development of new drugs in the stage of preclinical research and clinical trials, and the mechanism of action, implementation possibility, and clinical effects of these therapies are discussed. By summarizing the latest scientific research results, in this review, we attempt to paint a panorama of the mechanism of adenosine action in tumor immunotherapy, with the aim to provide a solid theoretical basis and practical guidance for subsequent research and clinical application, ultimately promoting the development of more accurate and efficient tumor immunotherapy.
Collapse
Affiliation(s)
- Liang Shan
- Department of Pharmacy, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Anhui Medical University), Hefei, Anhui, 230000, China
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, Anhui, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230032, China
- The Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui Province, 230032, China
| | - Mingxu Gong
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, Anhui, 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230032, China
- The Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui Province, 230032, China
| | - Dandan Zhai
- Department of Pharmacy, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Anhui Medical University), Hefei, Anhui, 230000, China
| | - Xiangyun Meng
- Department of Pharmacy, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Anhui Medical University), Hefei, Anhui, 230000, China
| | - Jianjun Liu
- Department of Pharmacy, The Second People's Hospital of Hefei (Hefei Hospital Affiliated to Anhui Medical University), Hefei, Anhui, 230000, China.
| | - Xiongwen Lv
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, Anhui, 230032, China.
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, 230032, China.
- The Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui Province, 230032, China.
| |
Collapse
|
3
|
Qin J, Tang N, Huang D, Zhang Z, Lu G, Chen Y, Huang X. Integrated proteomic network analysis reveals PTPRC as a central hub protein orchestrating co-expression modules and metabolic dysregulation in renal carcinoma: PTPRC protein molecular action. Int J Biol Macromol 2025; 311:144056. [PMID: 40345297 DOI: 10.1016/j.ijbiomac.2025.144056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/11/2025]
Abstract
The occurrence of renal carcinoma is closely related to a variety of molecular mechanisms and metabolic disorders. PTPRC (protein tyrosine phosphatase receptor C), as an important regulatory protein, was studied to reveal the role of PTPRC in renal carcinoma through comprehensive proteomic network analysis, especially its core position in the coordination of co-expression modules and metabolic disorders. This study was the first to download and process multiple publicly available renal cancer transcriptome data to conduct differential gene expression analysis across datasets. Functional enrichment and disease ontology analysis were performed on the transcriptome of renal cancer, and weighted gene co-expression network (WGCNA) was constructed. The results showed that comprehensive principal component analysis revealed significant differences in the transcriptome of renal cancer, and functional annotation revealed specific pathways associated with renal cancer. WGCNA analysis identified tumor-associated co-expression modules, while multi-omics analysis further identified core regulatory networks including PTPRC. As a central hub protein, PTPRC plays an important coordinating role in the co-expression module and metabolic dysregulation of renal carcinoma. This discovery provides a new perspective for understanding the molecular mechanism of kidney cancer.
Collapse
Affiliation(s)
- Junkai Qin
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Ning Tang
- Hepatobiliary Surgery Division, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang 524045, China
| | - Dongjun Huang
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Zhifu Zhang
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Guoping Lu
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China
| | - Yuanbo Chen
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China.
| | - Xijian Huang
- Department of Urology, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning 530001, China.
| |
Collapse
|
4
|
Wang J, Song Y, Tan X, Wang T, Shi Y, Xu X, Du J, Yu Z, Song B. Targeting PIM1 by Bruceine D attenuates skin fibrosis via myofibroblast ferroptosis. Redox Biol 2025; 82:103619. [PMID: 40168881 PMCID: PMC11993190 DOI: 10.1016/j.redox.2025.103619] [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: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025] Open
Abstract
Skin pan-fibrosis diseases-such as hypertrophic scar (HS), keloid scar (KS), and systemic sclerosis (SSc)-pose significant threats to patients' health and quality of life. In this study, the authors conducted both in vivo and in vitro experiments and discovered that the serine/threonine kinase PIM1 is upregulated in the myofibroblasts of human HS, KS, and SSc tissues, as well as in various animal models of skin fibrosis. Overexpression of PIM1 enhanced the profibrotic phenotypes of human hypertrophic scar fibroblasts (HSFs), which serve as key effector cells in the pathogenesis of skin pan-fibrosis diseases. Through high-throughput screening and subsequent laboratory assays, we identified the small molecule Bruceine D (BD) as a direct binder of PIM1. BD promoted ferroptosis in HSFs by selectively suppressing the PIM1-KEAP1-NRF2 pathway through augmented degradation of PIM1. In various in vivo models-including a hypertrophic scar mouse model, a rabbit ear hypertrophic scar model, and a bleomycin (BLM)-induced skin fibrosis mouse model-BD effectively attenuated fibrotic phenotypes. Collectively, these findings demonstrate that PIM1 serves as a common biomarker and therapeutic target for skin pan-fibrosis diseases. BD mitigates skin fibrosis by activating ferroptosis via PIM1 inhibition, highlighting its great translational potential and high promise to be developed to a clinical drug in treating these conditions, especially those with abnormally elevated PIM1 expression.
Collapse
Affiliation(s)
- Jianzhang Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yajuan Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoying Tan
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - Tong Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Shi
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xingbo Xu
- Clinic for Cardiology and Pulmonology, University Medical Center Göttingen, Göttingen, 37075, Germany.
| | - Juan Du
- Department of Dermatology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Zhou Yu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Baoqiang Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
5
|
Lyu B, Gou W, Xu F, Chen L, Wang Z, Ren Z, Liu G, Li Y, Hou W. Target Discovery Driven by Chemical Biology and Computational Biology. CHEM REC 2025; 25:e202400182. [PMID: 39811950 DOI: 10.1002/tcr.202400182] [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: 09/09/2024] [Revised: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Target identification is crucial for drug screening and development because it can reveal the mechanism of drug action and ensure the reliability and accuracy of the results. Chemical biology, an interdisciplinary field combining chemistry and biology, can assist in this process by studying the interactions between active molecular compounds and proteins and their physiological effects. It can also help predict potential drug targets or candidates, develop new biomarker assays and diagnostic reagents, and evaluate the selectivity and range of active compounds to reduce the risk of off-target effects. Chemical biology can achieve these goals using techniques such as changing protein thermal stability, enzyme sensitivity, and molecular structure and applying probes, isotope labeling and mass spectrometry. Concurrently, computational biology employs a diverse array of computational models to predict drug targets. This approach also offers innovative avenues for repurposing existing drugs. In this paper, we review the reported chemical biology and computational biology techniques for identifying different types of targets that can provide valuable insights for drug target discovery.
Collapse
Affiliation(s)
- Bohai Lyu
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Wenfeng Gou
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Feifei Xu
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Leyuan Chen
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Zhiyun Wang
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Zhonghao Ren
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Gaiting Liu
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yiliang Li
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Wenbin Hou
- Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| |
Collapse
|
6
|
Wang Y, Wang J, Chen L, Zhou W, He H, Chen X, Wang H. Protective effect of Pinacidil on hypoxic-reoxygenated cardiomyocytes in vitro and in vivo via HIF-1α/HRE pathway. PLoS One 2025; 20:e0318859. [PMID: 39992983 PMCID: PMC11849820 DOI: 10.1371/journal.pone.0318859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Cardiomyocyte hypoxia-reoxygenation (HR) is considered as a major cause of heart failure. Pinacidil is a classic ATP sensitive potassium channel opener and plays a crucial role in cardiomyocyte HR injuries. However, the specific mechanism is poorly understood. We established HR rat model and introduced 5-Hydroxydecanoate (5-HD), N-(2-Mercaptopropionyl)-glycine (MPG), and Dimethylethylenediylglycine (DMOG) to investigate the protection of Pinacidil (P) on cardiomyocyte. HE staining, electron microscopy and JC-1 staining were used to observe mitochondrial structure and mitochondrial membrane potential (MMP). Reactive oxygen species (ROS), hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor A (VEGF-A), heme oxygenase-1 (HO-1), and induced nitric oxide synthase (iNOS) were analyzed in this study. Network pharmacology analysis and auto-docking were used to predict the possible target of Pinadicil under cardiomyocyte HR condition. The integrity of mitochondrial structure and MMP were effectively promoted in P and MPG+DMOG + P groups. ROS was significantly increased after HR, treatment with P or MPG+DMOG + P, the content of ROS was increased. The expressions of HIF-1α, VEGF-A, HO-1 and iNOS were significantly increased in P and MPG+DMOG + P groups compared with HR group. Docking results confirmed that prolyl hydroxylase (PHD) was the most possible target for unsaturated binding with Pinacidil guanidine. Altogether, these data indicate that Pinacidil up-regulated and activated HIF-1α protein to protect caridomyocytes against HR injuries and the mechanism may be related to Pinacidil guanidine binding to PHD.
Collapse
Affiliation(s)
- Ying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiaqi Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Liangen Chen
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wenjing Zhou
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Haifeng He
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiyuan Chen
- School of Anesthesiology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Haiying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
7
|
Yang Y, Liu M, Dong X, Bai J, Shi W, Zhu Q, Liu J, Wang Z, Yi L, Yin X, Ni J, Qu C. Naringin Suppresses CoCl 2-Induced Ferroptosis in ARPE-19 Cells. Antioxidants (Basel) 2025; 14:236. [PMID: 40002420 PMCID: PMC11852185 DOI: 10.3390/antiox14020236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxic damage to retinal pigment epithelial (RPE) cells and subsequent neovascularization are key factors in the pathogenesis of branch retinal vein occlusion (BRVO). Naringin (NG), a naturally occurring flavanone glycoside, has demonstrated significant antioxidant and anti-neovascular activities. However, the regulatory effects and mechanisms of NG on ferroptosis in BRVO are yet to be explored. Our study aimed to investigate the protective effects of NG on RPE cells under hypoxic stress and to elucidate the underlying molecular mechanisms. Our findings revealed that NG significantly reduced cytotoxicity induced by cobaltous chloride (CoCl2) and also inhibited vascular proliferation in the retina, thereby attenuating choroidal neovascularization. NG pretreatment largely countered the overproduction of reactive oxygen species (ROS) and malondialdehyde (MDA) triggered by hypoxic damage, while also restoring levels of the antioxidants glutathione (GSH) and superoxide dismutase (SOD). Furthermore, NG pretreatment significantly activated the expression of hypoxia-inducible factor-1 alpha (HIF-1α) and its downstream heme oxygenase-1 (HO-1) and NADPH dehydrogenase (NQO1). In conclusion, NG not only inhibits neovascularization but also alleviates inflammation in RPE cells by modulating the HO-1/GPX4 pathway to inhibit ferroptosis. These findings highlight the potential of NG as a promising therapeutic agent for the treatment of BRVO.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jian Ni
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| | - Changhai Qu
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| |
Collapse
|
8
|
Shi S, Ou X, Liu C, Wen H, Ke J. Research progress of HIF-1a on immunotherapy outcomes in immune vascular microenvironment. Front Immunol 2025; 16:1549276. [PMID: 39981236 PMCID: PMC11839635 DOI: 10.3389/fimmu.2025.1549276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
The hypoxia-inducible factor-1α (HIF-1α) plays a key role in facilitating the adaptation of cells to hypoxia, profoundly influencing the immune vascular microenvironment (IVM) and immunotherapy outcomes. HIF-1α-mediated tumor hypoxia drives angiogenesis, immune suppression, and extracellular matrix remodeling, creating an environment that promotes tumor progression and resistance to immunotherapies. HIF-1α regulates critical pathways, including the expression of vascular endothelial growth factor and immune checkpoint upregulation, leading to tumor-infiltrating lymphocyte dysfunction and recruitment of immunosuppressive cells like regulatory T cells and myeloid-derived suppressor cells. These alterations reduce the efficacy of checkpoint inhibitors and other immunotherapies. Recent studies highlight therapeutic strategies that target HIF-1α, such as the use of pharmacological inhibitors, gene editing techniques, and hypoxia-modulating treatments, which show promise in enhancing responses to immunotherapy. This review explores the molecular mechanisms of action of HIF-1α in IVM, its impact on immunotherapy resistance, as well as potential interventions, emphasizing the need for innovative approaches to circumvent hypoxia-driven immunosuppression in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Jiang Ke
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Wang S, Han J, Wang Z, Liu X, Wang C, Nisar MF, Pan L, Xu K. Targeted Therapy of Tumors and Cancer Stem Cells based on Oxidant-regulated Redox Pathway and its Mechanism. Curr Comput Aided Drug Des 2025; 21:425-440. [PMID: 38818918 DOI: 10.2174/0115734099299174240522115944] [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: 12/17/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 06/01/2024]
Abstract
A malignant tumor is a frequent and common disease that severely threatens human health. Many mechanisms, such as cell signaling pathway, anti-apoptosis mechanism, cell stemness, metabolism, and cell phenotype, have been studied to explain the reasons for chemotherapy, radioresistance, and tumor recurrences in antitumor treatment. Cancer stem cells (CSCs) are important tumor cell subclasses that can potentially organize and regulate stem cell properties. Growing evidence suggests that CSCs can initiate tumors and constitute a significant factor in metastasis, recurrence, and treatment resistance. The inability to completely target and remove CSCs is a considerable obstacle in tumor treatment. Therefore, drugs and therapeutic strategies that can effectively intervene with CSCs are essential for the treatment of different tumor types. However, the current strategies and efficacy of targeted elimination of CSCs are very limited. Oxidative stress has been recognized to play a crucial role in cancer pathophysiology. Moreover, reactive oxygen species (ROS) production and imbalance of the built-in cellular antioxidant defense system are hallmarks of tumor and cancer etiology. The current paper will focus on the regulation and mechanism behind oxidative stress in tumors and cancer stem cells and its tumor therapy applications. Additionally, the article discusses the role of CSCs in causing tumor treatment resistance and recurrence based on a redox perspective. The study also emphasizes that targeted modulation of oxidative stress in CSCs has great potential in tumor therapy, providing novel prospects for tumor therapy.
Collapse
Affiliation(s)
- Shunshun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Juanjuan Han
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zijun Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xianqiong Liu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chunli Wang
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, 63100, Pakistan
| | - Lianhong Pan
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, 400030, China
| | - Kang Xu
- Hubei Provincial Engineering Technology Research Center for Chinese Medicine Processing, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| |
Collapse
|
10
|
He SJ, Li J, Zhou JC, Yang ZY, Liu X, Ge YW. Chemical proteomics accelerates the target discovery of natural products. Biochem Pharmacol 2024; 230:116609. [PMID: 39510194 DOI: 10.1016/j.bcp.2024.116609] [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: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
More than half of the global novel drugs are directly or indirectly derived from natural products (NPs) because of their better selectivity towards proteins. Traditional medicines perform multiple bioactivities through various NPs binding to drug targets, which highlights the opportunities of target discovery for drug development. However, detecting the binding relationship between NPs and targets remains challenging. Chemical proteomics, an interdisciplinary field of chemistry, proteomics, biology, and bioinformatics, has emerged as a potential approach for uncovering drug-target interactions. This review summarizes the principles and characteristics of the current widely applied chemical proteomic technologies, while delving into their latest applications in the target discovery of natural medicine. These endeavours demonstrate the potential of chemical proteomics for target discovery to supply dependable methodologies for the target elucidation of NPs.
Collapse
Affiliation(s)
- Shu-Jie He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jun Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie-Chun Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhi-You Yang
- College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Institute of Nutrition and Marine Drugs, Guangdong Ocean University, Zhanjiang, China
| | - Xi Liu
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yue-Wei Ge
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
11
|
Hop NQ, Son NT. The quassinoids bruceines A-M: pharmacology, mechanism of action, synthetic advance, and pharmacokinetics-a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9417-9433. [PMID: 38985315 DOI: 10.1007/s00210-024-03281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Bruceines A-L are among the quassinoid representatives found in the medicinal plant Brucea javanica (L.). An overview of their pharmacological activities is still unknown. The given research deals with highlights in their pharmacological result, molecular mechanism of action, synthetic progress, and pharmacokinetics. From previous evidence, bruceine derivatives are potential agents for anticancer treatments, as well as they are appropriate to treat inflammation, diabetes, and parasitic infections, and protect the neurons, kidneys, and lungs. Cytokine inhibitions, oxidative stress responses, and various signaling pathways, such as MAPK (mitogen-activated protein kinase) and NF-κB (nuclear factor-kappa B), have been proposed as the underlying mechanisms of action. Synthetic approaches to synthesize new derivatives with enhancement activities are based on free hydroxyl group modifications. Bruceines seem to be promptly absorbed by both oral and intravenous administrations, but their bioavailability is not high (less than 6%). Pre-clinical and clinical studies to prove their anticancer potential and other activities are urgent. Structural modifications, nano-combinations, and synergistic effects are necessary.
Collapse
Affiliation(s)
- Nguyen Quang Hop
- Faculty of Chemistry, Hanoi Pedagogical University 2 (HPU2), 32 Nguyen Van Linh, Xuanhoa, Phucyen, Vinhphuc, Vietnam
| | - Ninh The Son
- Graduate University of Science and Technology, VAST, 18 Hoang Quoc Viet, Caugiay, Hanoi, Vietnam.
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi, Vietnam.
| |
Collapse
|
12
|
Ma S, Meng G, Liu T, You J, He R, Zhao X, Cui Y. The Wnt signaling pathway in hepatocellular carcinoma: Regulatory mechanisms and therapeutic prospects. Biomed Pharmacother 2024; 180:117508. [PMID: 39362068 DOI: 10.1016/j.biopha.2024.117508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/26/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor that arises from hepatocytes. Multiple signaling pathways play a regulatory role in the occurrence and development of HCC, with the Wnt signaling pathway being one of the primary regulatory pathways. In normal hepatocytes, the Wnt signaling pathway maintains cell regeneration and organ development. However, when aberrant activated, the Wnt pathway is closely associated with invasion, cancer stem cells(CSCs), drug resistance, and immune evasion in HCC. Among these factors, the development of drug resistance is one of the most important factors affecting the efficacy of HCC treatment. These mechanisms form the basis for tumor cell adaptation and evolution within the body, enabling continuous changes in tumor cells, resistance to drugs and immune system attacks, leading to metastasis and recurrence. In recent years, there have been numerous new discoveries regarding these mechanisms. An increasing number of drugs targeting the Wnt signaling pathway have been developed, with some already entering clinical trials. Therefore, this review encompasses the latest research on the role of the Wnt signaling pathway in the onset and progression of HCC, as well as advancements in its therapeutic strategies.
Collapse
Affiliation(s)
- Shihui Ma
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Guorui Meng
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Tong Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Junqi You
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Risheng He
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Xudong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Yunfu Cui
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China.
| |
Collapse
|
13
|
Bao Z, Yang M, Guo Y, Ge Q, Zhang H. MTFR2 accelerates hepatocellular carcinoma mediated by metabolic reprogramming via the Akt signaling pathway. Cell Signal 2024; 123:111366. [PMID: 39182591 DOI: 10.1016/j.cellsig.2024.111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Metabolic reprogramming has recently been identified as a hallmark of malignancies. The shift from oxidative phosphorylation to glycolysis in hepatocellular carcinoma (HCC) meets the demands of rapid cell growth and provides a microenvironment for tumor progression. This study sought to uncover the function and mechanism of MTFR2 in the metabolic reprogramming of HCC. Elevated MTFR2 expression was associated with poor patient prognosis. Downregulation of MTFR2 blocked malignant behaviors, epithelial-to-mesenchymal transition (EMT), and glycolysis in HCC cells. Nuclear transcription factor Y subunit gamma (NFYC) was also associated with poor patient prognosis, and NFYC bound to the promoter of MTFR2 to activate transcription and promote Akt signaling. The repressive effects of NFYC knockdown on EMT and glycolysis in HCC cells were compromised by MTFR2 overexpression, elicited through the activation of the Akt signaling. Knockdown of NFYC slowed the growth and intrahepatic metastasis in vivo, which was reversed by MTFR2 overexpression. In conclusion, our work shows that activation of MTFR2 by the transcription factor NFYC promotes Akt signaling, thereby potentiating metabolic reprogramming in HCC development. Targeting the NFYC/MTFR2/Akt axis may represent a therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Zhongming Bao
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Ming Yang
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Yunhu Guo
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China
| | - Qi Ge
- School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239000, Anhui, PR China.
| | - Huaguo Zhang
- Department of Hepatobiliary Surgery, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huaiyin 223300, Jiangsu, PR China.
| |
Collapse
|
14
|
Gu WJ, Liu XX, Shen YW, Gong YT, Chen YL, Lin J, Lu D, Zhang LJ, Chen HZ, Jin Y, Zhan ZJ, Zhang WD, Jin JM, Luan X. TRIM4 enhances small-molecule-induced neddylated-degradation of CORO1A for triple negative breast cancer therapy. Theranostics 2024; 14:7023-7041. [PMID: 39629122 PMCID: PMC11610137 DOI: 10.7150/thno.97662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/13/2024] [Indexed: 12/06/2024] Open
Abstract
Background: As a critical member of the Coronin family, Coronin 1A (CORO1A) plays a crucial role in the progression of triple-negative breast cancer (TNBC). However, CORO1A is typically considered "undruggable" due to its smooth surface and complex protein-protein interactions (PPIs). Molecular glues have emerged as one of the most effective strategies to rapidly degrade such "undruggable" targets. Neddylation, an emerging approach, has shown promise in targeting pathogenic proteins for degradation through the NEDD8 pathway, making the degradation of CORO1A an attractive pharmacological strategy. Methods: A phenotypic drug screening strategy coupled with multi-omics approaches was utilized to rapidly identify a molecular glue degrader for CORO1A and to uncover the associated mechanisms. The Omics and Text-based Target Enrichment and Ranking (OTTER) tools, co-immunoprecipitation (Co-IP) assay, mass spectrometry, and the separation of phases-based protein interaction reporter (SPPIER) method were employed to explore the interaction between Aurovertin B (AB) and CORO1A via TRIM4. The pharmacological effects of AB were assessed using TNBC patient-derived organoids (PDOs) and 3D bioprinting models. Results: We identified AB as a previously undisclosed molecular glue that significantly promotes the neddylation and proteasomal degradation of CORO1A via TRIM4, an atypical E3 ligase. Notably, the degradation of CORO1A markedly inhibited various cellular processes and exerted robust antitumor effects in TNBC PDOs and 3D bioprinting models. Conclusions: Our findings underscore the critical role of CORO1A in TNBC and lay a crucial foundation for the development of innovative drugs based on molecular glue technology.
Collapse
Affiliation(s)
- Wen-Jie Gu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Xia Liu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi-Wen Shen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi-Ting Gong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi-Li Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dong Lu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi Jin
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education; Yunnan Provincial Center for Research & Development of Natural Products; School of Chemical Science and Technology, Yunnan University, Kunming 650091, China
| | - Zha-Jun Zhan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100700, China
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Jin-Mei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
15
|
Pan X, Jiang S, Zhang X, Wang Z, Wang X, Cao L, Xiao W. Recent strategies in target identification of natural products: Exploring applications in chronic inflammation and beyond. Br J Pharmacol 2024. [PMID: 39428703 DOI: 10.1111/bph.17356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/01/2024] [Accepted: 08/25/2024] [Indexed: 10/22/2024] Open
Abstract
Natural products are a treasure trove for drug discovery, especially in the areas of infection, inflammation and cancer, due to their diverse bioactivities and complex, and varied structures. Chronic inflammation is closely related to many diseases, including complex diseases such as cancer and neurodegeneration. Improving target identification for natural products contributes to elucidating their mechanism of action and clinical progress. It also facilitates the discovery of novel druggable targets and the elimination of undesirable ones, thereby significantly enhancing the productivity of drug discovery and development. Moreover, the rise of polypharmacological strategies, considered promising for the treatment of complex diseases, will further increase the demand for target deconvolution. This review underscores strategies for identifying natural product targets (NPs) in the context of chronic inflammation over the past 5 years. These strategies encompass computational methodologies for early target discovery and the anticipation of compound binding sites, proteomics-driven approaches for target delineation and experimental biology techniques for target validation and comprehensive mechanistic exploration.
Collapse
Affiliation(s)
- Xian Pan
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Shan Jiang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Xinzhuang Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Zhenzhong Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| | - Xin Wang
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Liang Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Xiao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
- Jiangsu Kanion Pharmaceutical Co Ltd, Jiangning Industrial City, Economic and Technological Development Zone of Lianyungang, Lianyungang, China
| |
Collapse
|
16
|
Deng J, Zhou J, Jiang B. Advances in the role of membrane-bound transcription factors in carcinogenesis and therapy. Discov Oncol 2024; 15:559. [PMID: 39404930 PMCID: PMC11480308 DOI: 10.1007/s12672-024-01414-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Protein shuttling between the cytoplasm and nucleus is a unique phenomenon in eukaryotic organisms, integral to various cellular functions. Membrane-bound transcription factors (MTFs), a specialized class of nucleocytoplasmic shuttling proteins, are anchored to the cell membrane and enter the nucleus upon ligand binding to exert their transcriptional regulatory functions. MTFs are crucial in cellular signal transduction, and aberrant nucleocytoplasmic shuttling of MTFs is closely associated with tumor initiation, progression, and resistance to anticancer therapies. Studies have demonstrated that MTFs, such as human epidermal growth factor receptor (HER), fibroblast growth factor receptor (FGFR), β-catenin, Notch, insulin-like growth factor 1 receptor (IGF-1R), and insulin receptor (IR), play critical roles in tumorigenesis and cancer progression. Targeted therapies developed against HERs and FGFRs, among these MTFs, have yielded significant success in cancer treatment. However, the development of drug resistance remains a major challenge. As research on MTFs progress, it is anticipated that additional MTF-targeted therapies will be developed to enhance cancer treatment. In this review, we summarized recent advancements in the study of MTFs and their roles in carcinogenesis and therapy, aiming to provide valuable insights into the potential of targeting MTF pathways for the reseach of therapeutic strategies.
Collapse
Affiliation(s)
- JiaLi Deng
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
- Department of Clinical Laboratory, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Jie Zhou
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
- Department of Clinical Laboratory, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China
| | - BinYuan Jiang
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China.
- Department of Clinical Laboratory, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, 410004, China.
| |
Collapse
|
17
|
Li SN, Ran RY, Chen J, Liu MC, Dang YM, Lin H. Angiogenesis in heterotopic ossification: From mechanisms to clinical significance. Life Sci 2024; 351:122779. [PMID: 38851421 DOI: 10.1016/j.lfs.2024.122779] [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: 12/28/2023] [Revised: 03/21/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
Heterotopic ossification (HO) refers to the formation of pathologic bone in nonskeletal tissues (including muscles, tendons or other soft tissues). HO typically occurs after a severe injury and can occur in any part of the body. HO lesions are highly vascularized. Angiogenesis, which is the formation of new blood vessels, plays an important role in the pathophysiology of HO. Surgical resection is considered an effective treatment for HO. However, it is difficult to completely remove new vessels, which can lead to the recurrence of HO and is often accompanied by significant problems such as intraoperative hemorrhage, demonstrating the important role of angiogenesis in HO. Here, we broadly summarize the current understanding of how angiogenesis contributes to HO; in particular, we focus on new insights into the cellular and signaling mechanisms underlying HO angiogenesis. We also review the development and current challenges associated with antiangiogenic therapy for HO.
Collapse
Affiliation(s)
- Sai-Nan Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; First Clinical School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Ruo-Yue Ran
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; First Clinical School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Jie Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Meng-Chao Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yan-Miao Dang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Hui Lin
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
18
|
Huang Y, Xiang P, Chen Y, Pan Q, Yuan K. Alantolactone facilitates ferroptosis in non-small cell lung cancer through promoting FTH1 ubiquitination and degradation. Chem Biol Drug Des 2024; 104:e14560. [PMID: 39175059 DOI: 10.1111/cbdd.14560] [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: 03/20/2024] [Revised: 04/26/2024] [Accepted: 05/20/2024] [Indexed: 08/24/2024]
Abstract
Alantolactone (ALT), a natural sesquiterpene lactone from Inula helenium L., demonstrates potent antitumor activity in various human cancers, notably non-small cell lung cancer (NSCLC). Despite its recognized efficacy, the precise mechanisms of action remain elusive. Our study aimed to elucidate ALT's impact on NSCLC. Our findings suggested that ALT triggered apoptosis both in vitro and in vivo, underscoring its anticancer potential. Interestingly, the ferroptosis inhibitor (Fer-1), rather than necrostatin-1 (Nec-1) or Z-VAD-FMK, rescued ALT-induced cell death, implicating ferroptosis as pivotal. Subsequent analyses revealed ferroptosis as the primary mechanism underlying ALT-induced NSCLC cell death, supported by markers including ROS accumulation, MDA elevation, GSH depletion, Fe2+ generation, and GPX4 reduction. Through DARTS/MS proteomics, we identified FTH1 as the target of ALT-induced ferroptosis. Immunoblotting confirmed ALT's inhibition of FTH1 protein expression and accelerated its degradation in NSCLC cells. Immunoprecipitation assays demonstrated increased FTH1 ubiquitination induced by ALT. Additionally, ALT induced ferroptosis and facilitated Fe2+ accumulation via FTH1 ubiquitination. Importantly, ALT displayed potent antitumor effects in a subcutaneous xenograft model in BALB/c-nu/nu nude mice by enhancing ferroptosis. In summary, ALT induced ferroptosis by promoting intracellular Fe2+ accumulation through accelerated FTH1 degradation, highlighting its potential as an antitumor agent targeting ferroptosis.
Collapse
Affiliation(s)
- Yijiao Huang
- Department of Oncology, Wuxi No.2 Chinese Medicine Hospital, Wuxi, Jiangsu, China
- Department of Oncology, Wuxi Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| | - Pei Xiang
- Department of Oncology, Wuxi No.2 Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Yuanyuan Chen
- Department of Oncology, Wuxi No.2 Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Qi Pan
- Department of Oncology, Wuxi No.2 Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Kemiao Yuan
- Department of Oncology, Wuxi No.2 Chinese Medicine Hospital, Wuxi, Jiangsu, China
- Department of Oncology, Wuxi Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, China
| |
Collapse
|
19
|
Wan M, Yu Q, Xu F, You LX, Liang X, Kang Ren K, Zhou J. Novel hypoxia-induced HIF-1αactivation in asthma pathogenesis. Respir Res 2024; 25:287. [PMID: 39061007 PMCID: PMC11282634 DOI: 10.1186/s12931-024-02869-0] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Asthma's complexity, marked by airway inflammation and remodeling, is influenced by hypoxic conditions. This study focuses on the role of Hypoxia-Inducible Factor-1 Alpha (HIF-1α) and P53 ubiquitination in asthma exacerbation. METHODS High-throughput sequencing and bioinformatics were used to identify genes associated with asthma progression, with an emphasis on GO and KEGG pathway analyses. An asthma mouse model was developed, and airway smooth muscle cells (ASMCs) were isolated to create an in vitro hypoxia model. Cell viability, proliferation, migration, and apoptosis were assessed, along with ELISA and Hematoxylin and Eosin (H&E) staining. RESULTS A notable increase in HIF-1α was observed in both in vivo and in vitro asthma models. HIF-1α upregulation enhanced ASMCs' viability, proliferation, and migration, while reducing apoptosis, primarily via the promotion of P53 ubiquitination through MDM2. In vivo studies showed increased inflammatory cell infiltration and airway structural changes, which were mitigated by the inhibitor IDF-11,774. CONCLUSION The study highlights the critical role of the HIF-1α-MDM2-P53 axis in asthma, suggesting its potential as a target for therapeutic interventions. The findings indicate that modulating this pathway could offer new avenues for treating the complex respiratory disorder of asthma.
Collapse
Affiliation(s)
- Mengzhi Wan
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Qi Yu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Fei Xu
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Lu Xia You
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Xiao Liang
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Kang Kang Ren
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China
| | - Jing Zhou
- Department of Respiratory Emergency and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province, 330006, PR China.
| |
Collapse
|
20
|
Wang T, Zhu G, Wang B, Hu M, Gong C, Tan K, Jiang L, Zhu X, Geng Y, Li L. Activation of Hypoxia Inducible Factor-1 Alpha-Mediated DNA Methylation Enzymes (DNMT3a and TET2) Under Hypoxic Conditions Regulates S100A6 Transcription to Promote Lung Cancer Cell Growth and Metastasis. Antioxid Redox Signal 2024; 41:138-151. [PMID: 38299557 DOI: 10.1089/ars.2023.0397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Aims: This research was aimed at investigating the effects of hypoxia inducible factor-1 alpha (HIF-1α)-mediated DNA methylation enzymes (ten-eleven translocase-2 [TET2] and DNA methyltransferase-3a [DNMT3a]) under hypoxic conditions on S100A6 transcription, thereby promoting the growth and metastasis of lung cancer cells. Methods: The expression of HIF-1α or S100A6 in lung cancer cells was interfered with under normoxic and hypoxic conditions, and the cell proliferative, migratory, and invasive properties were assessed. The mechanism of HIF-1α-regulated TET2 and DNMT3 effects on S100A6 transcription under hypoxic conditions was further investigated. Results: Functionally, S100A6 over-expression promoted lung cancer cell proliferation and metastasis. S100A6 over-expression reversed the inhibitory effects of HIF-1α interference on the proliferation and metastasis of lung cancer cells. S100A6 was induced to express in an HIF-1α-dependent manner under hypoxic conditions, and silencing S100A6 or HIF-1α suppressed lung cancer cell proliferation and metastasis under hypoxic conditions. Further, The Cancer Genome Atlas-lung adenocarcinoma database analysis revealed that S100A6 mRNA levels had a negative correlation with methylation levels. Mechanistically, CpG hypomethylation status in the S100A6 promoter hypoxia response element had an association with HIF-1α induction. TET2 was enriched in S100A6 promoter region of lung cancer cells under hypoxic conditions, whereas DNMT3a enrichment was reduced in S100A6 promoter region. HIF-1α-mediated S100A6 activation was linked to DNMT3a-associated epigenetic inactivation and TET2 activation. Innovation: The activation of HIF-1α-mediated DNA methylation enzymes under hypoxic conditions regulated S100A6 transcription, thereby promoting lung cancer cell growth and metastasis. Conclusion: In lung cancer progression, hypoxia-induced factor HIF-1α combined with DNA methylation modifications co-regulates S100A6 transcriptional activation and promotes lung cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Tengfei Wang
- The Department of Thoracic Surgery; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Genbao Zhu
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Bo Wang
- The Department of Thoracic Surgery; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Mengxue Hu
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Chen Gong
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - La Jiang
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Xiaohong Zhu
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Yuliu Geng
- The Department of Thoracic Surgery; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Lili Li
- General Clinical Research Center; Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
21
|
Zhu SL, Qi M, Chen MT, Lin JP, Huang HF, Deng LJ, Zhou XW. A novel DDIT3 activator dehydroevodiamine effectively inhibits tumor growth and tumor cell stemness in pancreatic cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155377. [PMID: 38503154 DOI: 10.1016/j.phymed.2024.155377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The existence of pancreatic cancer stem cells (PCSCs) results in limited survival benefits from current treatment options. There is a scarcity of effective agents for treating pancreatic cancer patients. Dehydroevodiamine (DeHE), a quinazoline alkaloid isolated from the traditional Chinese herb Evodiae fructus, exhibited potent inhibition of pancreatic ductal adenocarcinoma (PDAC) cell proliferation and tumor growth both in vitro and in vivo. METHODS The cytotoxic effect of DeHE on PDAC cells was assessed using CCK-8 and colony formation assays. The antitumor efficacy of DeHE were appraised in human PANC-1 xenograft mouse model. Sphere formation assay and flow cytometry were employed to quantify the tumor stemness. RNA-Seq analysis, drug affinity responsive target stability assay (DARTS), and RNA interference transfection were conducted to elucidate potential signaling pathways. Western blotting and immunohistochemistry were utilized to assess protein expression levels. RESULTS DeHE effectively inhibited PDAC cell proliferation and tumor growth in vitro and in vivo, and exhibited a better safety profile compared to the clinical drug gemcitabine (GEM). DeHE inhibited PCSCs, as evidenced by its suppression of self-renewal capabilities of PCSCs, reduced the proportion of ALDH+ cells and downregulated stemness-associated proteins (Nanog, Sox-2, and Oct-4) both in vitro and in vivo. Furthermore, there is potential involvement of DDIT3 and its downstream DDIT3/TRIB3/AKT/mTOR pathway in the suppression of stemness characteristics within DeHE-treated PDAC cells. Additionally, results from the DARTS assay indicated that DeHE interacts with DDIT3, safeguarding it against degradation mediated by pronase. Notably, the inhibitory capabilities of DeHE on PDAC cell proliferation and tumor stemness were partially restored by siDDIT3 or the AKT activator SC-79. CONCLUSION In summary, our study has identified DeHE, a novel antitumor natural product, as an activator of DDIT3 with the ability to suppress the AKT/mTOR pathway. This pathway is intricately linked to tumor cell proliferation and stemness characteristics in PDAC. These findings suggest that DeHE holds potential as a promising candidate for the development of innovative anticancer therapeutics.
Collapse
Affiliation(s)
- Su-Li Zhu
- Department of Biochemistry and Pharmacology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Ming Qi
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Mei-Ting Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, PR China
| | - Jia-Peng Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Hai-Fu Huang
- Internal Medicine-Oncology, Shenzhen Hospital of Guangzhou University of Traditional Chinese Medicine, PR China
| | - Li-Juan Deng
- Guangzhou Key Laboratory of Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, PR China.
| | - Xing-Wang Zhou
- Department of Biochemistry and Pharmacology, Sun Yat-Sen University Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China.
| |
Collapse
|
22
|
Saikia L, Gogoi B, Sen S, Tonk RK, Kumar D, Dutta PP. The recent update and advancements of natural products in targeting the Wnt/β-Catenin pathway for cancer prevention and therapeutics. Med Oncol 2024; 41:164. [PMID: 38816663 DOI: 10.1007/s12032-024-02387-5] [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: 03/13/2024] [Accepted: 04/19/2024] [Indexed: 06/01/2024]
Abstract
The Wnt/β-Catenin pathway (Wnt/β-CatP) is implicated in accelerating carcinogenesis and cancer progression, contributing to increased morbidity and treatment resistance. Even though it holds promise as a focus for cancer treatment, its intricate nature and diverse physiological effects pose significant challenges. Recent years have witnessed significant advancements in this domain, with numerous natural products demonstrating promising preclinical anti-tumor effects and identified as inhibitors of the Wnt/β-CatP through various upstream and downstream mechanisms. This study provides a comprehensive overview of the current landscape of Wnt/β-Cat-targeted cancer therapy, examining the impact of natural products on Wnt/β-Cat signaling in both cancer prevention and therapeutic contexts. A comprehensive search was conducted on scientific databases like SciFinder, PubMed, and Google Scholar to retrieve relevant literature on Wnt-signaling, natural products, β-Catenin (β-Cat), and cancer from 2020 to January 2024. As per the analysis of the relevant reference within the specified period, it has been noted that a total of 58 phytoconstituents, predominantly phenolics, followed by triterpenoids and several other classes, along with a limited number of plant extracts, have exhibited activity targeting the Wnt/β-CatP. Most β-Cat regulating modulators restrict cancer cell development by suppressing β-Cat expression, facilitating proteasomal degradation, and inhibiting nuclear translocation. Multiple approaches have been devised to block the activity of β-Cat in cancer therapy, a key factor in cancer progression, leading to the discovery of various Wnt/β-CatP regulators. However, their exploration remains limited, necessitating further research using clinical models for potential clinical use in cancer prevention and therapeutics.
Collapse
Affiliation(s)
- Lunasmrita Saikia
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India
| | - Bhaskarjyoti Gogoi
- Department of Biotechnology, The Assam Royal Global University, Guwahati, Assam, 781035, India
| | - Saikat Sen
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India
| | - Rajiv K Tonk
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India.
| |
Collapse
|
23
|
Cui Z, Li C, Liu W, Sun M, Deng S, Cao J, Yang H, Chen P. Scutellarin activates IDH1 to exert antitumor effects in hepatocellular carcinoma progression. Cell Death Dis 2024; 15:267. [PMID: 38622131 PMCID: PMC11018852 DOI: 10.1038/s41419-024-06625-6] [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: 12/22/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Isochlorate dehydrogenase 1 (IDH1) is an important metabolic enzyme for the production of α-ketoglutarate (α-KG), which has antitumor effects and is considered to have potential antitumor effects. The activation of IDH1 as a pathway for the development of anticancer drugs has not been attempted. We demonstrated that IDH1 can limit glycolysis in hepatocellular carcinoma (HCC) cells to activate the tumor immune microenvironment. In addition, through proteomic microarray analysis, we identified a natural small molecule, scutellarin (Scu), which activates IDH1 and inhibits the growth of HCC cells. By selectively modifying Cys297, Scu promotes IDH1 active dimer formation and increases α-KG production, leading to ubiquitination and degradation of HIF1a. The loss of HIF1a further leads to the inhibition of glycolysis in HCC cells. The activation of IDH1 by Scu can significantly increase the level of α-KG in tumor tissue, downregulate the HIF1a signaling pathway, and activate the tumor immune microenvironment in vivo. This study demonstrated the inhibitory effect of IDH1-α-KG-HIF1a on the growth of HCC cells and evaluated the inhibitory effect of Scu, the first IDH1 small molecule agonist, which provides a reference for cancer immunotherapy involving activated IDH1.
Collapse
Affiliation(s)
- Zhao Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Wei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Mo Sun
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing, China.
| |
Collapse
|
24
|
Tang W, Hu Y, Tu K, Gong Z, Zhu M, Yang T, Sarwar A, Dai B, Zhang D, Zhan Y, Zhang Y. Targeting Trop2 by Bruceine D suppresses breast cancer metastasis by blocking Trop2/β-catenin positive feedback loop. J Adv Res 2024; 58:193-210. [PMID: 37271476 PMCID: PMC10982870 DOI: 10.1016/j.jare.2023.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/28/2023] [Accepted: 05/28/2023] [Indexed: 06/06/2023] Open
Abstract
INTRODUCTION Tumor-associated calcium signal transducer 2 (Trop2) has been used as a transport gate for cytotoxic agents into cells in antibody-drug conjugate designs because of its expression in a wide range of solid tumors. However, the specific role of Trop2 itself in breast cancer progression remains unclear and small molecules targeting Trop2 have not yet been reported. OBJECTIVES To screen small molecules targeting Trop2, and to reveal its pharmacological effects and the molecular mechanisms of action. METHODS Small molecule targeting Trop2 was identified by cell membrane chromatography, and validated by cellular thermal shift assay and point mutation analyses. We investigated the pharmacological effects of Trop2 inhibitor using RNA-seq, human foreskin fibroblast (HFF)-derived extracellular matrix (ECM), Matrigel drop invasion assays, colony-forming assay, xenograft tumor model, 4T1 orthotopic metastasis model and 4T1 experimental metastasis model. The molecular mechanism was determined using immunoprecipitation, mass spectrometry, immunofluorescence, immunohistochemistry and Western blotting. RESULTS Here we identified Bruceine D (BD) as the inhibitor of Trop2, and demonstrated anti-metastasis effects of BD in breast cancer. Notably, Lys307 and Glu310 residues of Trop2 acted as critical sites for BD binding. Mechanistically, BD suppressed Trop2-induced cancer metastasis by blocking the formation of Trop2/β-catenin positive loop, in which the Trop2/β-catenin complex prevented β-catenin from being degraded via the ubiquitin-proteosome pathway. Destabilized β-catenin caused by BD reduced nucleus translocation, leading to the reduction of transcription of Trop2, the reversal of epithelial-mesenchymal transition (EMT) process, and the inhibition of ECM remodeling, further inhibiting cancer metastasis. Additionally, the inhibitory effects of BD on lung metastatic colonization and the beneficial effects of BD on prolongation of survival were validated in 4T1 experimental metastasis model. CONCLUSIONS These results support the tumor-promoting role of Trop2 in breast cancer by stabilizing β-catenin in Trop2/β-catenin positive loop, and suggest Bruceine D as a promising candidate for Trop2 inhibition.
Collapse
Affiliation(s)
- Wenjuan Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Yu Hu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Kaihui Tu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Zhengyan Gong
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Tianfeng Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Ammar Sarwar
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Dongdong Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China
| | - Yingzhuan Zhan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi'an 710061, China.
| |
Collapse
|
25
|
Gajos-Michniewicz A, Czyz M. WNT/β-catenin signaling in hepatocellular carcinoma: The aberrant activation, pathogenic roles, and therapeutic opportunities. Genes Dis 2024; 11:727-746. [PMID: 37692481 PMCID: PMC10491942 DOI: 10.1016/j.gendis.2023.02.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/28/2022] [Accepted: 02/14/2023] [Indexed: 09/12/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a liver cancer, highly heterogeneous both at the histopathological and molecular levels. It arises from hepatocytes as the result of the accumulation of numerous genomic alterations in various signaling pathways, including canonical WNT/β-catenin, AKT/mTOR, MAPK pathways as well as signaling associated with telomere maintenance, p53/cell cycle regulation, epigenetic modifiers, and oxidative stress. The role of WNT/β-catenin signaling in liver homeostasis and regeneration is well established, whereas in development and progression of HCC is extensively studied. Herein, we review recent advances in our understanding of how WNT/β-catenin signaling facilitates the HCC development, acquisition of stemness features, metastasis, and resistance to treatment. We outline genetic and epigenetic alterations that lead to activated WNT/β-catenin signaling in HCC. We discuss the pivotal roles of CTNNB1 mutations, aberrantly expressed non-coding RNAs and complexity of crosstalk between WNT/β-catenin signaling and other signaling pathways as challenging or advantageous aspects of therapy development and molecular stratification of HCC patients for treatment.
Collapse
Affiliation(s)
- Anna Gajos-Michniewicz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz 92-215, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz 92-215, Poland
| |
Collapse
|
26
|
Yan G, Xiao Q, Zhao J, Chen H, Xu Y, Tan M, Peng L. Brucea javanica derived exosome-like nanovesicles deliver miRNAs for cancer therapy. J Control Release 2024; 367:425-440. [PMID: 38295998 DOI: 10.1016/j.jconrel.2024.01.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/05/2024]
Abstract
Triple-negative breast cancer (TNBC) is characterized by complex heterogeneity, high recurrence and metastasis rates, and short overall survival, owing to the lack of endocrine and targeted receptors, which necessitates chemotherapy as the major treatment regimen. Exosome-like nanovesicles derived from medicinal plants have shown great potential as novel biotherapeutics for cancer therapy by delivering their incorporated nucleic acids, especially microRNAs (miRNAs), to mammalian cells. In this study, we isolated exosome-like nanovesicles derived from B. javanica (BF-Exos) and investigated their influence and underlying molecular mechanisms in TNBC. We found that BF-Exos delivered 10 functional miRNAs to 4T1 cells, significantly retarding the growth and metastasis of 4T1 cells by regulating the PI3K/Akt/mTOR signaling pathway and promoting ROS/caspase-mediated apoptosis. Moreover, BF-Exos were shown to inhibit the secretion of vascular endothelial growth factor, contributing to anti-angiogenesis in the tumor microenvironment. In vivo, BF-Exos inhibited tumor growth, metastasis, and angiogenesis in breast tumor mouse models, while maintaining high biosafety. Overall, BF-Exos are considered promising nanoplatforms for the delivery of medicinal plant-derived nucleic acids, with great potential to be developed into novel biotherapeutics for the treatment of TNBC.
Collapse
Affiliation(s)
- Ge Yan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Qiyao Xiao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Jingyu Zhao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Haoran Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Yang Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Minhong Tan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Lihua Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, PR China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, PR China.
| |
Collapse
|
27
|
Chen X, Li H. Bruceine D and Narclasine inhibit the proliferation of breast cancer cells and the prediction of potential drug targets. PLoS One 2024; 19:e0297203. [PMID: 38215156 PMCID: PMC10786365 DOI: 10.1371/journal.pone.0297203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/31/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Breast cancer is one of the most common female malignancies. This study explored the underlying mechanism through which the two plant compounds (Brucaine D and Narclasine) inhibited the proliferation of breast cancer cells. OBJECTIVE The purpose of this study was to explore the effect of Brucaine D and Narclasine on breast cancer development and their potential drug targets. METHODS GSE85871 dataset containing 212 samples and the hallmark gene set "h.all.v2023.1.Hs.symbols.gmt" were downloaded from the Gene Expression Omnibus (GEO) database and the Molecular Signatures Database (MSigDB) database, respectively. Principal component analysis (PCA) was applied to classify clusters showing similar gene expression pattern. Single sample gene set enrichment analysis (ssGSEA) was used to calculate the hallmark score for different drug treatment groups. The expressions of genes related to angiogenesis, glycolysis and cell cycle were detected. Protein-protein interaction (PPI) network analysis was performed to study the interaction of the hub genes. Then, HERB database was employed to identify potential target genes for Narclasine and Bruceine D. Finally, in vitro experiments were conducted to validate partial drug-target pair. RESULTS PCA analysis showed that the significant changes in gene expression patterns took place in 6 drugs treatment groups (Narciclasine, Bruceine D, Japonicone A, 1beta-hydroxyalatolactone, Britanin, and four mixture drugs) in comparison to the remaining drug treatment groups. The ssGSEA pathway enrichment analysis demonstrated that Narciclasine and Bruceine treatments had similar enriched pathways, for instance, suppressed pathways related to angiogenesis, Glycolysis, and cell cycle, etc.. Further gene expression analysis confirmed that Narciclasine and Bruceine had a strong ability to inhibit these cell cycle genes, and that MYC, CHEK2, MELK, CDK4 and EZH2 were closely interacted with each other in the PPI analysis. Drug target prediction revealed that Androgen Receptor (AR) and Estrogen Receptor 1 (ESR1) were the targets for Bruceine D, and Cytochrome P450 3A4 enzyme (CYP3A4) was the target for Narciclasine. Cell experiments also confirmed the connections between Narciclasine and CYP3A4. CONCLUSION The present study uncovered that Narciclasine and Bruceine D could inhibit the growth of breast cancer and also predicted the potential targets for these two drugs, providing a new therapeutic direction for breast cancer patients.
Collapse
Affiliation(s)
- Xinhao Chen
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hua Li
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
28
|
Chen H, Han X, Zhang Y, Wang K, Liu D, Hu Z, Wang J. Bruceine D suppresses CAF-promoted TNBC metastasis under TNF-α stimulation by inhibiting Notch1-Jagged1/NF-κB(p65) signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:154928. [PMID: 38043386 DOI: 10.1016/j.phymed.2023.154928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) has a poor prognosis because of its high degree of malignancy and the lack of effective treatment options. Cancer-associated fibroblasts (CAFs) comprise the most abundant stromal cells in the tumor microenvironment (TME), leading to functional impairments and facilitating tumor metastasis. Excessive TNF-α further promotes cross-talk between different cells in TME. Therefore, there is an urgent need to develop more effective therapies and potential drugs that target the key factors that promote TNBC metastasis. PURPOSE The study aimed to evaluate the efficacy of Bruceine D, an active compound derived from the Chinese herb Brucea javanica, in inhibiting metastasis and elucidate the underlying mechanism of action in TNBC. METHODS In vitro, the clonogenic and the Transwell assays were used to assess the effects of Bruceine D on the proliferation, migration and invasion abilities of co-cultured CAFs and MDA-MB-231 (4T1) cells under TNF-α stimulation. TNF-α, IL-6, CXCL12, TGF-β1, and MMP9 levels in the supernatant of co-cultured cells were determined using ELISA. Western blotting was utilized to detect the expression levels of proteins related to the Notch1-Jagged1/NF-κB(p65) pathway. In vivo, the anti-tumor growth and anti-metastatic effectiveness of Bruceine D was evaluated by determining tumor weight, number of metastatic lesions, and pathological changes in the tumor and lung/liver tissues. The inhibitory effect of Bruceine D on α-SMA+ CAFs activation and CAF-medicated extracellular matrix remodeling was accessed using immunohistochemistry, immunofluorescence, and Masson and Sirius Red staining. The expression levels of Notch1, Jagged1 and p-NF-κB(p65) proteins in the primary tumors were measured by immunohistochemistry and western blotting. RESULTS In vitro, Bruceine D significantly inhibited the migration and invasion of co-cultured CAFs and MDA-MB-231 (4T1) cells under TNF-α stimulation, reduced the expression of tumor-promoting and matrix-remodeling cytokines secreted by CAFs, and hindered the mutual activation of Notch1-Jagged1 and NF-κB(p65). In vivo, Bruceine D significantly suppressed tumor growth and the formation of lung and liver metastases by decreasing TNF-α stimulated α-SMA+ CAFs activation, collagen fibers, MMPs production, and inhibited Notch1-Jagged1/NF-κB(p65) signaling in TNBC-bearing mice. CONCLUSION Bruceine D effectively weakened the "tumor-CAF-inflammation" network by inhibiting the mutual activation of Notch1-Jagged1 and NF-κB(p65) and thereby suppressed TNBC metastasis. This study first explored that Bruceine D disrupted the cross-talk between CAFs and tumor cells under TNF-α stimulation to inhibit the metastasis of TNBC, and highlighted the potential of Bruceine D as therapeutic agent for suppressing tumor metastasis.
Collapse
Affiliation(s)
- Han Chen
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, 750004, Yinchuan, China; The First Affiliated Hospital of Xi'an Medical University, 48 Fenghao West Road, Lianhu District, 710082, Xian, China
| | - Xue Han
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, 750004, Yinchuan, China
| | - Yue Zhang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, 750004, Yinchuan, China
| | - Ke Wang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, 750004, Yinchuan, China
| | - Da Liu
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, 750004, Yinchuan, China
| | - Zhiqiang Hu
- Oncology Hospital, General Hospital of Ningxia Medical University, 804 Shengli Street, 750004, Yinchuan, China.
| | - Jing Wang
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, 750004, Yinchuan, China; Key Laboratory of Ningxia Minority Medicine Modernization, Ministry of Education, 1160 Shengli Street, 750004, Yinchuan, China.
| |
Collapse
|
29
|
Yang Y, Li P, Li X, Zhu Y, Guo X. Brucine D restrains colorectal cancer tumorigenesis and autophagy by downregulating circ_0068464. Chem Biol Drug Des 2024; 103:e14407. [PMID: 38040413 DOI: 10.1111/cbdd.14407] [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: 08/23/2023] [Revised: 10/18/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023]
Abstract
Bruceine D (BD) from Brucea javanica (L) exerts an antitumor effect in several human cancers. At present, it has not been reported whether BD inhibits the malignancy of colorectal cancer (CRC) cells. Therefore, investigating the role and regulatory mechanisms of BD in CRC is the main thrust of this study. Effect of BD on CRC cell viability, proliferation, apoptosis, invasion, and autophagy was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide, 5-ethynyl-2'-deoxyuridine, flow cytometry, transwell invasion, and western blotting assays. Expression changes of has_circ_0068464 (circ_0068464) were detected using real time quantitative polymerase chain reaction. The molecular mechanisms related to circ_0068464 were predicted through online prediction websites Starbase 2.0, circinteractome, and CircBank and validated using dual-luciferase reporter and RNA pull-down assays. The tumorigenic ability of BD and circ_0068464 on CRC was confirmed by xenograft experiments. The results showed that BD lessened CRC cell proliferation, invasion, autophagy, and prompted cell apoptosis. Circ_0068464 was overexpressed in CRC samples and cells. BD led to a significant reduction in circ_0068464 levels in cells of this carcinoma, but circ_0068464 overexpression partially rescued these effects urged by BD. Also, the combination of BD and circ_0068464 silencing decreased xenograft tumor growth compared to BD alone. Importantly, circ_0068464 could regulate ATG5 expression by functioning as a miR-520h molecular sponge. In conclusion, BD might suppress CRC growth by inhibiting the circ_0068464/miR-520h/ATG5 axis, providing a new perspective for the molecular pathogenesis of CRC and preliminarily indicating that BD may be a promising drug for CRC treatment.
Collapse
Affiliation(s)
- Yong Yang
- Department Anus & Intestine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peng Li
- Department Anus & Intestine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojia Li
- Department Anus & Intestine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Zhu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiutian Guo
- Department Anus & Intestine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
30
|
Wan J, Zhang Z, Wu C, Tian S, Zang Y, Jin G, Sun Q, Wang P, Luan X, Yang Y, Zhan X, Ye LL, Duan DD, Liu X, Zhang W. Astragaloside IV derivative HHQ16 ameliorates infarction-induced hypertrophy and heart failure through degradation of lncRNA4012/9456. Signal Transduct Target Ther 2023; 8:414. [PMID: 37857609 PMCID: PMC10587311 DOI: 10.1038/s41392-023-01660-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/10/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Reversing ventricular remodeling represents a promising treatment for the post-myocardial infarction (MI) heart failure (HF). Here, we report a novel small molecule HHQ16, an optimized derivative of astragaloside IV, which effectively reversed infarction-induced myocardial remodeling and improved cardiac function by directly acting on the cardiomyocyte to reverse hypertrophy. The effect of HHQ16 was associated with a strong inhibition of a newly discovered Egr2-affiliated transcript lnc9456 in the heart. While minimally expressed in normal mouse heart, lnc9456 was dramatically upregulated in the heart subjected to left anterior descending coronary artery ligation (LADL) and in cardiomyocytes subjected to hypertrophic stimulation. The critical role of lnc9456 in cardiomyocyte hypertrophy was confirmed by specific overexpression and knockout in vitro. A physical interaction between lnc9456 and G3BP2 increased NF-κB nuclear translocation, triggering hypertrophy-related cascades. HHQ16 physically bound to lnc9456 with a high-affinity and induced its degradation. Cardiomyocyte-specific lnc9456 overexpression induced, but knockout prevented LADL-induced, cardiac hypertrophy and dysfunction. HHQ16 reversed the effect of lnc9456 overexpression while lost its protective role when lnc9456 was deleted, further confirming lnc9456 as the bona fide target of HHQ16. We further identified the human ortholog of lnc9456, also an Egr2-affiliated transcript, lnc4012. Similarly, lnc4012 was significantly upregulated in hypertrophied failing hearts of patients with dilated cardiomyopathy. HHQ16 also specifically bound to lnc4012 and caused its degradation and antagonized its hypertrophic effects. Targeted degradation of pathological increased lnc4012/lnc9456 by small molecules might serve as a novel promising strategy to regress infarction-induced cardiac hypertrophy and HF.
Collapse
Affiliation(s)
- Jingjing Wan
- School of Pharmacy, Second Military Medical University, Shanghai, PR China
| | - Zhen Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, PR China
| | - Chennan Wu
- School of Pharmacy, Second Military Medical University, Shanghai, PR China
| | - Saisai Tian
- School of Pharmacy, Second Military Medical University, Shanghai, PR China
| | - Yibei Zang
- School of Pharmacy, Second Military Medical University, Shanghai, PR China
| | - Ge Jin
- School of Pharmacy, Second Military Medical University, Shanghai, PR China
| | - Qingyan Sun
- China Institute of Pharmaceutical Industry, Shanghai, PR China
| | - Pin Wang
- Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, PR China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yili Yang
- China Regional Research Centre, International Centre of Genetic Engineering & Biotechnology, Taizhou, PR China
| | - Xuelin Zhan
- China Regional Research Centre, International Centre of Genetic Engineering & Biotechnology, Taizhou, PR China
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, PR China
| | - Lingyu Linda Ye
- Center for Phenomics of Traditional Chinese Medicine, Hospital of Traditional Chinese Medicine Affiliated to Southwest Medical University, Southwest Medical University, Luzhou, PR China
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, Hospital of Traditional Chinese Medicine Affiliated to Southwest Medical University, Southwest Medical University, Luzhou, PR China.
- Key Laboratory of Autoimmune Diseases and Precision Medicine, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, PR China.
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, PR China.
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, PR China.
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
31
|
Hu L, Gao M, Jiang H, Zhuang L, Jiang Y, Xie S, Zhang H, Wang Q, Chen Q. Triptolide inhibits epithelial ovarian tumor growth by blocking the hedgehog/Gli pathway. Aging (Albany NY) 2023; 15:11131-11151. [PMID: 37851362 PMCID: PMC10637820 DOI: 10.18632/aging.205110] [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: 03/23/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023]
Abstract
Epithelial ovarian cancer (EOC), the most predominant subtype of ovarian cancer (OC), involves poor prognosis and exhibits high aggression. Triptolide (TPL), like other Chinese herbs, has historically played a significant role in modern medicine. The screening system based on Gli-dependent luciferase reporter activity assessed the effects of over 800 natural medicinal materials on hedgehog (Hh) signaling pathway activity and discovered that TPL had an excellent inhibitory effect on Hh signaling pathway activity. However, the significance and mechanism of TPL involvement in regulating the Hh pathway have not been well explored. Thus, this work aimed to understand better how TPL affects the Hh pathway activity, which, in turn, influences the biological behavior of EOC. Our findings observed that Smo agonist SAG-induced EOC cell proliferation, migration, and invasion were drastically reversed by TPL in a concentration-dependent pattern. Further evidence suggested that TPL promotes the degradation of Gli1 and Gli2 to inhibit the activity of the Hh signaling pathway by relying on Gli1 and Gli2 ubiquitination. Our in vivo studies also confirmed that TPL could significantly inhibit the tumor growth of EOC. Taken together, our results revealed that one of the antitumor mechanisms of TPL was the targeted inhibition of the Hh/Gli pathway.
Collapse
Affiliation(s)
- Lanyan Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Mai Gao
- Huankui Academy of Nanchang University, Nanchang 330036, Jiangxi, P.R. China
| | - Huifu Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Lingling Zhuang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Ying Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Siqi Xie
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qian Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qi Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
32
|
Zhang J, Han H, Wang L, Wang W, Yang M, Qin Y. Overcoming the therapeutic resistance of hepatomas by targeting the tumor microenvironment. Front Oncol 2022; 12:988956. [DOI: 10.3389/fonc.2022.988956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for the majority of primary liver cancers and is the third leading cause of cancer-related mortality worldwide. Multifactorial drug resistance is regarded as the major cause of treatment failure in HCC. Accumulating evidence shows that the constituents of the tumor microenvironment (TME), including cancer-associated fibroblasts, tumor vasculature, immune cells, physical factors, cytokines, and exosomes may explain the therapeutic resistance mechanisms in HCC. In recent years, anti-angiogenic drugs and immune checkpoint inhibitors have shown satisfactory results in HCC patients. However, due to enhanced communication between the tumor and TME, the effect of heterogeneity of the microenvironment on therapeutic resistance is particularly complicated, which suggests a more challenging research direction. In addition, it has been reported that the three-dimensional (3D) organoid model derived from patient biopsies is more intuitive to fully understand the role of the TME in acquired resistance. Therefore, in this review, we have focused not only on the mechanisms and targets of therapeutic resistance related to the contents of the TME in HCC but also provide a comprehensive description of 3D models and how they contribute to the exploration of HCC therapies.
Collapse
|
33
|
Cui Y, Li C, Sang F, Cao W, Qin Z, Zhang P. Natural products targeting glycolytic signaling pathways-an updated review on anti-cancer therapy. Front Pharmacol 2022; 13:1035882. [PMID: 36339566 PMCID: PMC9631946 DOI: 10.3389/fphar.2022.1035882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Glycolysis is a complex metabolic process that occurs to convert glucose into pyruvate to produce energy for living cells. Normal cells oxidized pyruvate into adenosine triphosphate and carbon dioxide in the presence of oxygen in mitochondria while cancer cells preferentially metabolize pyruvate to lactate even in the presence of oxygen in order to maintain a slightly acidic micro-environment of PH 6.5 and 6.9, which is beneficial for cancer cell growth and metastasis. Therefore targeting glycolytic signaling pathways provided new strategy for anti-cancer therapy. Natural products are important sources for the treatment of diseases with a variety of pharmacologic activities. Accumulated studies suggested that natural products exhibited remarkable anti-cancer properties both in vitro and in vivo. Plenty of studies suggested natural products like flavonoids, terpenoids and quinones played anti-cancer properties via inhibiting glucose metabolism targets in glycolytic pathways. This study provided an updated overview of natural products controlling glycolytic pathways, which also provide insight into druggable mediators discovery targeting cancer glucose metabolism.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Chuang Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Zhuo Qin
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| |
Collapse
|
34
|
Cui Q, Ding W, Liu P, Luo B, Yang J, Lu W, Hu Y, Huang P, Wen S. Developing Bi-Gold Compound BGC2a to Target Mitochondria for the Elimination of Cancer Cells. Int J Mol Sci 2022; 23:ijms232012169. [PMID: 36293028 PMCID: PMC9602679 DOI: 10.3390/ijms232012169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 12/04/2022] Open
Abstract
Reactive oxygen species (ROS) homeostasis and mitochondrial metabolism are critical for the survival of cancer cells, including cancer stem cells (CSCs), which often cause drug resistance and cancer relapse. Auranofin is a mono-gold anti-rheumatic drug, and it has been repurposed as an anticancer agent working by the induction of both ROS increase and mitochondrial dysfunction. Hypothetically, increasing auranofin’s positive charges via incorporating more gold atoms to enhance its mitochondria-targeting capacity could enhance its anti-cancer efficacy. Hence, in this work, both mono-gold and bi-gold compounds were designed and evaluated to test our hypothesis. The results showed that bi-gold compounds generally suppressed cancer cells proliferation better than their mono-gold counterparts. The most potent compound, BGC2a, substantially inhibited the antioxidant enzyme TrxR and increased the cellular ROS. BGC2a induced cell apoptosis, which could not be reversed by the antioxidant agent vitamin C, implying that the ROS induced by TrxR inhibition might not be the decisive cause of cell death. As expected, a significant proportion of BGC2a accumulated within mitochondria, likely contributing to mitochondrial dysfunction, which was further confirmed by measuring oxygen consumption rate, mitochondrial membrane potential, and ATP production. Moreover, BGC2a inhibited colony formation and reduced stem-like side population (SP) cells of A549. Finally, the compound effectively suppressed the tumor growth of both A549 and PANC-1 xenografts. Our study showed that mitochondrial disturbance may be gold-based compounds’ major lethal factor in eradicating cancer cells, providing a new approach to developing potent gold-based anti-cancer drugs by increasing mitochondria-targeting capacity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peng Huang
- Correspondence: (P.H.); (S.W.); Tel.: +86-20-87343511 (P.H.); +86-20-87342283 (S.W.)
| | - Shijun Wen
- Correspondence: (P.H.); (S.W.); Tel.: +86-20-87343511 (P.H.); +86-20-87342283 (S.W.)
| |
Collapse
|
35
|
Yu F, Tan W, Chen Z, Shen X, Mo X, Mo X, He J, Deng Z, Wang J, Luo Z, Yang J. Nitidine chloride induces caspase 3/GSDME-dependent pyroptosis by inhibting PI3K/Akt pathway in lung cancer. Chin Med 2022; 17:115. [PMID: 36175965 PMCID: PMC9524076 DOI: 10.1186/s13020-022-00671-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/12/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND As the increasing mortality and incidence of lung cancer (LC), there is an urgent need to discover novel treatment agent. In this study, we aimed to investigate the anti-LC effects of nitidine chloride (NC), a small molecular compound extracted from Chinese herbal medicine, while detailing its underlying mechanisms. METHODS Cell viability was detected by MTT assays and five cell death inhibitors, including ferrostatin-1 (Fer-1), Z-VAD-FMK, necrostatin-1 (Nec-1), disulfiram (DSF) and IM-54 were used to explore the type of cell death induced by NC. The microscopic features of NC-induced pyroptosis were assessed by transmission electron microscopy (TEM) and the pyroptotic-related proteins such as caspase and gasdermin family, were examined by western blot. Network pharmacology was employed to predict the potential mechanisms of NC in lung cancer treatment. CETSA and DARTs were used to determine the activity of NC binding to targeted protein. Xenograft mice model was established to further investigate the inhibitory effect and mechanism of NC against LC. RESULTS The pyroptosis inhibitor (DSF) and apoptosis inhibitor (Z-VAD-FMK) but not IM-54, necrostatin-1, or Ferrostatin-1 rescued NC-induced cell death. Morphologically, H1688 and A549 cells treated with NC showed notably pyroptotic features, such as cell swelling and large bubbles emerging from the plasma membrane. Gasdermin E (GSDME) rather than GSDMC or GSDMD was cleaved in NC-treated H1688 and A549 cells with an increased cleavage of caspase 3. Combined with network pharmacology and molecule docking, PI3K/Akt signaling axis was predicted and was further verified by CETSA and DARTs assay. In addition, the activation of PI3K is able to rescue the pyroptosis induced by NC in vitro. In xenograft model of LC, NC significantly hindered the transduction of PI3K-AKT pathway, inducing pyroptosis of tumor. CONCLUSION Our data indicated that NC is a potential therapeutic agent for the treatment of LC via triggering GSDME-dependent pyroptosis.
Collapse
Affiliation(s)
- Fei Yu
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Weidan Tan
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Zhiquan Chen
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Xiaoju Shen
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Xiaoxiang Mo
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Xiaocheng Mo
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Jingchuan He
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Zhihua Deng
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Jie Wang
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Zhuo Luo
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| | - Jie Yang
- grid.256607.00000 0004 1798 2653Department of Pharmacology, School of Pharmacy, 406 Graduate School of Guangxi Medical University, Nanning, 530021 Guangxi People’s Republic of China
| |
Collapse
|
36
|
Hao S, Meng Q, Sun H, Li Y, Li Y, Gu L, Liu B, Zhang Y, Zhou H, Xu Z, Wang Y. The role of transketolase in human cancer progression and therapy. Biomed Pharmacother 2022; 154:113607. [PMID: 36030587 DOI: 10.1016/j.biopha.2022.113607] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/02/2022] Open
Abstract
Transketolase (TKT) is an enzyme that is ubiquitously expressed in all living organisms and has been identified as an important regulator of cancer. Recent studies have shown that the TKT family includes the TKT gene and two TKT-like (TKTL) genes; TKTL1 and TKTL2. TKT and TKTL1 have been reported to be involved in the regulation of multiple cancer-related events, such as cancer cell proliferation, metastasis, invasion, epithelial-mesenchymal transition, chemoradiotherapy resistance, and patient survival and prognosis. Therefore, TKT may be an ideal target for cancer treatment. More importantly, the levels of TKTL1 were detected using EDIM technology for the early detection of some malignancies, and TKTL1 was more sensitive and specific than traditional tumor markers. Detecting TKTL1 levels before and after surgery could be used to evaluate the surgery's effect. While targeted TKT suppresses cancer in multiple ways, in some cases, it has detrimental effects on the organism. In this review, we discuss the role of TKT in different tumors and the detailed mechanisms while evaluating its value and limitations in clinical applications. Therefore, this review provides a basis for the clinical application of targeted therapy for TKT in the future, and a strategy for subsequent cancer-related research.
Collapse
Affiliation(s)
- Shiming Hao
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yao Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Liting Gu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
37
|
Effect of Shenqi Fuzheng Injection on Leukopenia and T-cell Subsets in Patients with Non-small Cell Lung Cancer Undergoing Radiotherapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2832739. [PMID: 35966722 PMCID: PMC9374546 DOI: 10.1155/2022/2832739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/11/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
Purpose The aim of this study is to evaluate the effect of Shenqi fuzheng injection on leukopenia and T-cell subsets in patients with non-small cell lung cancer (NSCLC) undergoing radiotherapy. Methods A total of 124 patients with advanced NSCLC treated in the oncology department of our hospital from January 2017 to January 2019 were included and assigned at a ratio of 1 : 1 to receive conventional radiotherapy (control group, n = 62) or conventional radiotherapy plus Shenqi Fuzheng injection (study group, n = 62) via the random number table method. Results The study group showed a significantly higher objective response rate (ORR) and a lower incidence of leukopenia versus the control group (P < 0.05). After the treatment, Shenqi Fuzheng injection resulted in significantly lower levels of carcinoembryonic antigen (CEA) and neuron-specific enolase (NSE) in the study group versus conventional treatment given to the control group. After the treatment, the control group showed significantly decreased ratios of CD3+ T cells, CD4+ T cells, and CD4+/CD8+, and an increased ratio of CD8+ T cells, and significant differences when compared with the study group. The T-cell subsets of the patients in the study group showed no significant changes than those between the treatment. The median OS was 20.0 months in the control group and 23.5 months in the study group. The differences between the two groups in terms of OS did not come up to the statistical standard. Conclusion Shenqi Fuzheng injection for NSCLC patients undergoing radiotherapy elevates the number of white blood cells, regulates T-cell immune function, reduces tumor markers, and enhances clinical efficacy. Further clinical trials are, however, required prior to clinical promotion.
Collapse
|
38
|
A new strategy for the rapid identification and validation of direct toxicity targets of psoralen-induced hepatotoxicity. Toxicol Lett 2022; 363:11-26. [PMID: 35597499 DOI: 10.1016/j.toxlet.2022.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/14/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
The interaction between small-molecule compounds of traditional Chinese medicine and their direct targets is the molecular initiation event, which is the key factor for toxicity efficacy. Psoralen, an active component of Fructus Psoraleae, is toxic to the liver and has various pharmacological properties. Although the mechanism of psoralen-induced hepatotoxicity has been studied, the direct target of psoralen remains unclear. Thus, the aim of this study was to discover direct targets of psoralen. To this end, we initially used proteomics based on drug affinity responsive target stability (DARTS) technology to identify the direct targets of psoralen. Next, we used surface plasmon resonance (SPR) analysis and verified the affinity effect of the 'component-target protein'. This method combines molecular docking technology to explore binding sites between small molecules and proteins. SPR and molecular docking confirmed that psoralen and tyrosine-protein kinase ABL1 could be stably combined. Based on the above experimental results, ABL1 is a potential direct target of psoralen-induced hepatotoxicity. Finally, the targets Nrf2 and mTOR, which are closely related to the hepatotoxicity caused by psoralen, were predicted by integrating proteomics and network pharmacology. The direct target ABL1 is located upstream of Nrf2 and mTOR, Nrf2 can influence the expression of mTOR by affecting the level of reactive oxygen species. Immunofluorescence experiments and western blot results showed that psoralen could affect ROS levels and downstream Nrf2 and mTOR protein changes, whereas the ABL1 inhibitor imatinib and ABL1 agonist DPH could enhance or inhibit this effect. In summary, we speculated that when psoralen causes hepatotoxicity, it acts on the direct target ABL1, resulting in a decrease in Nrf2 expression, an increase in ROS levels and a reduction in mTOR expression, which may cause cell death. We developed a new strategy for predicting and validating the direct targets of psoralen. This strategy identified the toxic target, ABL1, and the potential toxic mechanism of psoralen.
Collapse
|