1
|
Wang D, Lv L, Du J, Tian K, Chen Y, Chen M. TRIM16 and PRC1 Are Involved in Pancreatic Cancer Progression and Targeted by Delphinidin. Chem Biol Drug Des 2024; 104:e70026. [PMID: 39635962 DOI: 10.1111/cbdd.70026] [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: 08/02/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Pancreatic cancer (PC) is the leading cause of cancer-related death worldwide, and new biomarkers, therapeutic targets, and candidate drugs are needed. In this work, three PC-related microarray datasets (GSE183795, GSE28735, and GSE62452) were analyzed. The differentially expressed genes (DEGs) of PC were obtained with the limma package in R. Weighted gene co-expression network analysis (WGCNA) and machine learning approaches were used to screen the hub genes. Kaplan-Meier plotter and receiver operating characteristic (ROC) curve analysis were utilized to assess the diagnostic efficacy of the hub genes. The binding ability between natural bioactive ingredients and hub proteins was evaluated by molecular docking and molecular dynamics simulation. CCK-8, flow cytometry, transwell, and western blot assays were used to analyze the viability, apoptosis, cell cycle progression, invasion, and pathway change of PC cells. Additionally, a nude mice model was used to evaluate the aggressive properties of PC cells in vivo. In this study, a total of 988 DEGs were identified, which were mainly enriched in cell adhesion and PI3K-Akt signaling pathway, and two core genes TRIM16 and PRC1 were further identified. The overall survival of patients with high expression of TRIM16 and PRC1 was shorter. Delphinidin (Del) had good binding affinity with both TRIM16 and PRC1, and Del could inhibit the viability, invasion, and metastasis of PC cells and induce cell apoptosis and G0/G1 phase arrest. In addition, Del could promote the activation of p53 and inhibit the activation of the PI3K/AKT signaling pathway in PC cells. In summary, TRIM16 and PRC1 are identified as prognostic biomarkers and therapeutic targets for PC, and Del has good binding affinity with them and may be a potential therapeutic agent for PC.
Collapse
Affiliation(s)
- Donghua Wang
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Long Lv
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Jinghu Du
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Kui Tian
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Manyu Chen
- Department of Coloproctological Surgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
2
|
Lin L, Liu Y, Tang R, Ding S, Lin H, Li H. Evodiamine: A Extremely Potential Drug Development Candidate of Alkaloids from Evodia rutaecarpa. Int J Nanomedicine 2024; 19:9843-9870. [PMID: 39345907 PMCID: PMC11430234 DOI: 10.2147/ijn.s459510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 07/23/2024] [Indexed: 10/01/2024] Open
Abstract
Evodiamine (EVO) is a tryptamine indole alkaloid and the main active ingredient in Evodia rutaecarpa. In recent years, the antitumor, cardioprotective, anti-inflammatory, and anti-Alzheimer's disease effects of EVO have been reported. EVO exerts antitumor effects by inhibiting tumor cell activity and proliferation, blocking the cell cycle, promoting apoptosis and autophagy, and inhibiting the formation of the tumor microvasculature. However, EVO has poor solubility and low bioavailability. Several derivatives with high antitumor activity have been discovered through the structural optimization of EVO, and new drug delivery systems have been developed to improve the solubility and bioavailability of EVO. Current research found that EVO could have toxic effects, such as hepatotoxicity, nephrotoxicity, and cardiac toxicity. This article reviews the pharmacological activity, derivatives, drug delivery systems, toxicity, and pharmacokinetics of EVO and provides research ideas and references for its further in-depth development and clinical applications.
Collapse
Affiliation(s)
- Longfei Lin
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Yuling Liu
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Ruying Tang
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Shilan Ding
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Hongmei Lin
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People's Republic of China
- National Medical Products Administration Key Laboratory for Research Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Hui Li
- Institute Chinese Materia Medica China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, People's Republic of China
| |
Collapse
|
3
|
Ren QL, Li XL, Tian T, Li S, Shi RY, Wang Q, Zhu Y, Wang M, Hu H, Liu JG. Application of Natural Medicinal Plants Active Ingredients in Oral Squamous Cell Carcinoma. Chin J Integr Med 2024; 30:852-864. [PMID: 38607612 DOI: 10.1007/s11655-024-3804-7] [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] [Accepted: 12/18/2023] [Indexed: 04/13/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant cancer of the head and neck, with high morbidity and mortality, ranking as the sixth most common cancer in the world. The treatment of OSCC is mainly radiotherapy, chemotherapy and surgery, however, the prognosis of patients is still poor and the recurrence rate is high. This paper reviews the range of effects of natural medicinal plant active ingredients (NMPAIs) on OSCC cancer, including the types of NMPAIs, anti-cancer mechanisms, involved signaling pathways, and clinical trials. The NMPAIs include terpenoids, phenols, flavonoids, glycosides, alkaloids, coumarins, and volatile oils. These active ingredients inhibit proliferation, induce apoptosis and autophagy, inhibit migration and invasion of OSCC cells, and regulate cancer immunity to exert anti-cancer effects. The mechanism involves signaling pathways such as mitogen-activated protein kinase, phosphatidylinositol 3 kinase/protein kinase B, nuclear factor kappa B, miR-22/WNT1/β-catenin and Nrf2/Keap1. Clinically, NMPAIs can inhibit the growth of OSCC, and the combined drug is more effective. Natural medicinal plants are promising candidates for the treatment of OSCC.
Collapse
Affiliation(s)
- Qun-Li Ren
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Xiao-Lan Li
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Tian Tian
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Shuang Li
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Rong-Yi Shi
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Qian Wang
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Yuan Zhu
- School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Miao Wang
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Huan Hu
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China
| | - Jian-Guo Liu
- School of Stomatology, Zunyi Medical University, Zunyi, Guizhou Province, 563000, China.
| |
Collapse
|
4
|
Zhou Z, Zhou Y, Zhang Z, Zhao M, Hu C, Yang L, Zhou X, Zhang X, Liu L, Shen T. Progress on the effects and underlying mechanisms of evodiamine in digestive system diseases, and its toxicity: A systematic review and meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155851. [PMID: 39018943 DOI: 10.1016/j.phymed.2024.155851] [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: 07/09/2023] [Revised: 01/15/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Evodiamine (EVO) is one of the primary components of Evodia rutaecarpa and has been found to have a positive therapeutic effect on various digestive system diseases. However, no systematic review has been conducted on the research progress and mechanisms of EVO in relation to digestive system diseases, and its toxicity. PURPOSE This study aimed to provide a reference for future research in this field. STUDY DESIGN A systematic review and meta-analysis of the research progress, mechanisms, and toxicity of EVO in the treatment of digestive system diseases. METHODS Five electronic databases were utilized to search for relevant experiments. We conducted a comprehensive review and meta-analysis of the pertinent literature following the guidelines of Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA). RESULTS EVO's animal experiments in digestive system diseases primarily focus on colorectal cancer, gastric ulcers, liver cancer, liver fibrosis, ulcerative colitis, colitis-associated cancer, and functional gastrointestinal disorders. EVO also has positive effects on pancreatic cancer, radiation enteritis, gastric cancer, tongue squamous cancer, hepatitis B, oral cancer, and esophageal cancer in vivo. EVO's in cellular experiments primarily focus on SGC7901, HT29, HCT-116, and HepG2 cells. EVO also exhibits positive effects on SW480, LoVo, BGC-823, AGS, COLO-205, MKN45, SMMC-7721, Bel-7402, QGY7-701, PANC-1, SW1990, BxPC-3, HSC4, MC3, HONE1, and CNE1 cells in vitro. The potential common pathways include TGF-β, PI3K-AKT, Wnt, ErbB, mTOR, MAPK, HIF-1, NOD-like receptor, NF-κB, VEGF, JAK-STAT, AMPK, Toll-like receptor, EGFR, Ras, TNF, AGE-RAGE, Relaxin, FoxO, IL-17, Hippo, and cAMP. The mechanisms of EVO on ulcerative colitis, gastric cancer, and HCT116 cells are still controversial in vivo. EVO may have a bidirectional regulatory effect on functional gastrointestinal disorders through calcium signaling. The mechanisms of EVO on HCT116, HT29, SW480, AGS, COLO-205, and SW1990 cells are still controversial in vitro. The question of whether EVO has obvious toxicity is controversial. CONCLUSION In both cellular and animal experiments, EVO has demonstrated positive impacts on digestive system diseases. Nevertheless, additional in vivo and in vitro research is required to confirm the beneficial effects and mechanisms of EVO on digestive system diseases, as well as its potential toxicity.
Collapse
Affiliation(s)
- Zubing Zhou
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China
| | - Yan Zhou
- South Sichuan Preschool Education College, Neijiang, China
| | - Zhongyi Zhang
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China
| | - Mei Zhao
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China
| | - Chao Hu
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China
| | - Lele Yang
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China
| | - Xin Zhou
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China
| | - Xiaobo Zhang
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China.
| | - Liyun Liu
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China.
| | - Tao Shen
- Chengdu University of Traditional Chinese Medicine, College of Basic Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu 611137, China; Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
5
|
Tian Z, Zhu L, Xie Y, Hu H, Ren Q, Liu J, Wang Q. The mechanism of high mobility group box-1 protein and its bidirectional regulation in tumors. BIOMOLECULES & BIOMEDICINE 2024; 24:477-485. [PMID: 37897664 PMCID: PMC11088895 DOI: 10.17305/bb.2023.9760] [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: 09/03/2023] [Revised: 10/01/2023] [Accepted: 10/26/2023] [Indexed: 10/30/2023]
Abstract
High-mobility group box-1 protein (HMGB1) is a nonhistone chromatin-related protein widely found in eukaryotic cells. It is involved in the transcription, replication, and repair of DNA to maintain nuclear homeostasis. It participates in cell growth, differentiation, and signal transduction. Recent studies showed that HMGB1 has a bidirectional regulatory effect on tumors by regulating TLR4/MYD88/NF-κB and RAGE/AMPK/mTOR signaling pathways. On the one hand, it is highly expressed in a variety of tumors, promoting tumor proliferation and invasion, while on the other hand, it induces autophagy and apoptosis of tumor cells and stimulates tumor-infiltrating lymphocytes to produce an anti-tumor immune response. At present, HMGB1 could be used as a target to regulate the drug resistance and prognostication in cancer. Clinical applications of HMGB1 in cancer need further in-depth studies.
Collapse
Affiliation(s)
- Zhongjia Tian
- The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Lin Zhu
- The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Yutong Xie
- The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Huan Hu
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qunli Ren
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Jianguo Liu
- The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Wang
- The Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
6
|
Lin LQ, Lv SY, Ren HZ, Li RR, Li L, Pang YQ, Wang J. Evodiamine inhibits EPRS expression to regulate glutamate metabolism and proliferation of oral squamous cell carcinoma cells. Kaohsiung J Med Sci 2024; 40:348-359. [PMID: 38243370 DOI: 10.1002/kjm2.12803] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024] Open
Abstract
The effects of evodiamine (EVO) on oral squamous cell carcinoma (OSCC) are not yet understood. Based on our earlier findings, we hypothesized that evodiamine may affect OSCC cell proliferation and glutamate metabolism by modulating the expression of EPRS (glutamyl-prolyl-tRNA synthetase 1). From GEPIA, we obtained EPRS expression data in patients with OSCC as well as survival prognosis data. An animal model using Cal27 cells in BALB/c nude mice was established. The expression of EPRS was assessed by immunofluorescence, Western blotting, and quantitative PCR. Glutamate measurements were performed to evaluate the impact of evodiamine on glutamate metabolism of Cal27 and SAS tumor cells. transient transfection techniques were used to knock down and modulate EPRS in these cells. EPRS is expressed at higher levels in OSCC than in normal tissues, and it predicts poor prognosis in patients. In a nude mouse xenograft model, evodiamine inhibited tumor growth and the expression of EPRS. Evodiamine impacted cell proliferation, glutamine metabolism, and EPRS expression on Cal27 and SAS cell lines. In EPRS knockdown cell lines, both cell proliferation and glutamine metabolism are suppressed. EPRS's overexpression partially restores evodiamine's inhibitory effects on cell proliferation and glutamine metabolism. This study provides crucial experimental evidence supporting the potential therapeutic application of evodiamine in treating OSCC. Evodiamine exhibits promising anti-tumor effects by targeting EPRS to regulate glutamate metabolism.
Collapse
Affiliation(s)
- Li-Qi Lin
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Si-Yi Lv
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Hao-Zhe Ren
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Rong-Rong Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Lin Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Yun-Qing Pang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
- Clinical Research Center for Oral Diseases, Lanzhou, Gansu Province, China
| | - Jing Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu, China
- Clinical Research Center for Oral Diseases, Lanzhou, Gansu Province, China
| |
Collapse
|
7
|
Fan A, Gao M, Tang X, Jiao M, Wang C, Wei Y, Gong Q, Zhong J. HMGB1/RAGE axis in tumor development: unraveling its significance. Front Oncol 2024; 14:1336191. [PMID: 38529373 PMCID: PMC10962444 DOI: 10.3389/fonc.2024.1336191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
High mobility group protein 1 (HMGB1) plays a complex role in tumor biology. When released into the extracellular space, it binds to the receptor for advanced glycation end products (RAGE) located on the cell membrane, playing an important role in tumor development by regulating a number of biological processes and signal pathways. In this review, we outline the multifaceted functions of the HMGB1/RAGE axis, which encompasses tumor cell proliferation, apoptosis, autophagy, metastasis, and angiogenesis. This axis is instrumental in tumor progression, promoting tumor cell proliferation, autophagy, metastasis, and angiogenesis while inhibiting apoptosis, through pivotal signaling pathways, including MAPK, NF-κB, PI3K/AKT, ERK, and STAT3. Notably, small molecules, such as miRNA-218, ethyl pyruvate (EP), and glycyrrhizin exhibit the ability to inhibit the HMGB1/RAGE axis, restraining tumor development. Therefore, a deeper understanding of the mechanisms of the HMGB1/RAGE axis in tumors is of great importance, and the development of inhibitors targeting this axis warrants further exploration.
Collapse
Affiliation(s)
- Anqi Fan
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Mengxiang Gao
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
8
|
Chen X, Liu Q, Wu E, Ma Z, Tuo B, Terai S, Li T, Liu X. The role of HMGB1 in digestive cancer. Biomed Pharmacother 2023; 167:115575. [PMID: 37757495 DOI: 10.1016/j.biopha.2023.115575] [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: 08/08/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023] Open
Abstract
High mobility group box protein B1 (HMGB1) belongs to the HMG family, is widely expressed in the nucleus of digestive mucosal epithelial cells, mesenchymal cells and immune cells, and binds to DNA to participate in genomic structural stability, mismatch repair and transcriptional regulation to maintain normal cellular activities. In the context of digestive inflammation and tumors, HMGB1 readily migrates into the extracellular matrix and binds to immune cell receptors to affect their function and differentiation, further promoting digestive tract tissue injury and tumor development. Notably, HMGB1 can also promote the antitumor immune response. Therefore, these seemingly opposing effects in tumors make targeted HMGB1 therapies important in digestive cancer. This review focuses on the role of HMGB1 in tumors and its effects on key pathways of digestive cancer and aims to provide new possibilities for targeted tumor therapy.
Collapse
Affiliation(s)
- Xiangqi Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Qian Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Enqing Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Shuji Terai
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
9
|
Green V, Baldwin L, England J, Marshall G, Frost L, Moore C, Greenman J. Head and Neck Squamous Cell Carcinoma Biopsies Maintained Ex Vivo on a Perfusion Device Show Gene Changes with Time and Clinically Relevant Doses of Irradiation. Cancers (Basel) 2023; 15:4575. [PMID: 37760543 PMCID: PMC10527562 DOI: 10.3390/cancers15184575] [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: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Advancements in 3-Dimensional (3D) culture models for studying disease have increased significantly over the last two decades, but fully understanding how these models represent in vivo still requires further investigation. The current study investigated differences in gene expression between a baseline sample and that maintained on a tissue-on-chip perfusion device for up to 96 h, with and without clinically-relevant doses of irradiation, to allow differentiation of model and treatment effects. Tumour tissue samples from 7 Head and Neck Squamous Cell Carcinomas (HNSCC) patients were sub-divided and either fixed immediately upon excision or maintained in a tissue-on-chip device for 48 and 96 h, with or without 2 Gray (Gy) or 10 Gy irradiation. Gene expression was measured using an nCounter® PanCancer Progression Panel. Differentially expressed genes between pre- and post-ex vivo culture, and control and irradiated samples were identified using nSolver software (version 4.0). The secretome from the tumour-on-chip was analysed for the presence of cytokines using a Proteome Profiler™ platform. Significant numbers of genes both increased (n = 6 and 64) and decreased (n = 18 and 58) in expression in the tissue maintained on-chip for 48 and 96 h, respectively, compared to fresh tissue; however, the irradiation schedule chosen did not induce significant changes in gene expression or cytokine secretion. Although HNSCC tissue maintained ex vivo shows a decrease in a large proportion of altered genes, 25% and 53% (48 and 96 h) do show increased expression, suggesting that the tissue remains functional. Irradiation of tumour tissue-on-chip needs to be conducted for longer time periods for specific gene changes to be observed, but we have shown, for the first time, the feasibility of using this perfusion platform for studying the genomic response of HNSCC tissue biopsies.
Collapse
Affiliation(s)
- Victoria Green
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| | - Lydia Baldwin
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| | - James England
- Department of Otorhinolaryngology, Head and Neck Surgery, Hull University Teaching Hospitals NHS Trust Hull, Hull HU16 5JQ, UK;
| | - Gayle Marshall
- Medicines Discovery Catapult Ltd., Alderley Park, Alderley Edge, Cheshire SK10 4TG, UK; (G.M.); (L.F.)
| | - Lucy Frost
- Medicines Discovery Catapult Ltd., Alderley Park, Alderley Edge, Cheshire SK10 4TG, UK; (G.M.); (L.F.)
| | - Craig Moore
- Medical Physics Service, Hull University Teaching Hospitals NHS Trust Hull, Hull HU16 5JQ, UK;
| | - John Greenman
- Centre for Biomedicine, Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK; (L.B.); (J.G.)
| |
Collapse
|
10
|
Plemmenos G, Tzimogianni V, Fili C, Piperi C. Contributing Role of High Mobility Group Box 1 Signaling in Oral Cancer Development and Therapy. Life (Basel) 2023; 13:1577. [PMID: 37511951 PMCID: PMC10381251 DOI: 10.3390/life13071577] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most frequent type of oral cancer of multifactorial origin, characterized by histological and clinical manifestations. To date, there are no specific biomarkers or treatment modalities available to efficiently manage this neoplasia, demanding further research on the molecular background of OSCC pathology. Elucidation of signal transduction pathways and associated molecules with differential expression and function in OSCC are expected to enhance the future development of molecular targeted therapies. Among signaling proteins with a potential functional role in OSCC, the High Mobility Group Box 1 (HMGB1) protein has stimulated scientific interest due to frequent upregulation, and implication in the progression of many types of head and neck cancer types. HMGB1 is a nuclear nonhistone protein and an extracellularly secreted cytokine that can interact with several signaling molecules implicated in the pathogenic pathways of OSCC. Binding of HMGB1 to specific receptors on OSCC cells such as the receptor of AGE (RAGE) and the toll-like receptor (TLR) has been shown to initiate several intercellular signaling cascades that can promote OSCC growth, invasion, and metastasis, indicating a potential target for patient prognosis and therapeutic approaches. The purpose of this review is to explore the functional role and associated signaling of HMGB1 in OSCC in order to reveal potential therapeutic targeting options.
Collapse
Affiliation(s)
- Grigorios Plemmenos
- School of Dentistry, National and Kapodistrian University of Athens, 2 Thivon Str, Goudi, 11527 Athens, Greece
| | - Valentini Tzimogianni
- Department of Biology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527 Athens, Greece
| | - Christina Fili
- Medicine and Surgery, Department of Pharmacy and Medicine, Sapienza Universita di Roma, Piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527 Athens, Greece
| |
Collapse
|
11
|
Zhi S, Li J, Kong X, Xie X, Zhang Q, Fang G. Insulin-like growth factor 2 mRNA binding protein 2 regulates proliferation, migration, and angiogenesis of keratinocytes by modulating heparanase stability. Bioengineered 2021; 12:11267-11276. [PMID: 34753397 PMCID: PMC8810085 DOI: 10.1080/21655979.2021.2002495] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Wound healing is related to proliferation, migration, and angiogenesis of keratinocytes. Insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) is an important N6-methyladenosine (m6A) reader, which is involved in multiple processes, including wound healing. However, the function and mechanism of IGF2BP2 in keratinocyte processes are largely uncertain. In the present study, expression levels of IGF2BP2 and heparanase (HPSE) were detected by quantitative reverse transcription polymerase chain reaction and western blotting assays. Cell proliferation was investigated by cell counting kit-8 (CCK-8) analysis. Cell migration was determined through wound healing assay. Angiogenesis was measured by tube formation assay and vascular endothelial growth factor (VEGF) level using enzyme linked immunosorbent assay (ELISA). The interaction between IGF2BP2 and HPSE was analyzed by RNA immunoprecipitation, pull-down and luciferase reporter analyses. The results showed that IGF2BP2 expression was enhanced in wound healing. IGF2BP2 downregulation constrained HaCaT cell proliferation, migration, and angiogenesis. IGF2BP2 knockdown decreased HPSE expression. IGF2BP2 could regulate HPSE stability by binding with 3ʹ untranslated region (UTR) of HPSE. HPSE upregulation attenuated silencing IGF2BP2-mediated suppression of proliferation, migration, and angiogenesis. As a conclusion, IGF2BP2 knockdown repressed proliferation, migration, and angiogenesis of HaCaT cells by decreasing HPSE stability.
Collapse
Affiliation(s)
- Shaomin Zhi
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Jun Li
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Xiao Kong
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Xuemei Xie
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Qiangli Zhang
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| | - Guoxiang Fang
- Department of Emergency, Xi'an No. 3 Hospital, Xi'an, Shaanxi, P.R. China
| |
Collapse
|