1
|
Rao H, An X, Qu X, Yu J, Xie J, Ke J, Liu Z, You L, Qiu Z, Tian L, Du W, Li W, Jia J, Liu D, Li S. SGLT2i delays c-Myc-induced HCC progression via targeting mTOR. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167805. [PMID: 40113049 DOI: 10.1016/j.bbadis.2025.167805] [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: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) stands as a primary malignant liver tumor characterized by metabolic reprogramming. The oncogene c-Myc exerts substantial influence by driving the transcription of numerous genes. Empagliflozin (EMPA), a sodium-glucose cotransporter-2 inhibitor (SGLT2i), is widely used in the treatment of type 2 diabetes and has recently attracted attention for its potential anti-cancer effects. This study aims to unravel the complex interplay among c-Myc, EMPA, and the mammalian target of rapamycin (mTOR) in HCC development and progression. METHODS HCC induction in mice utilized high-pressure hydrodynamic transfection of the c-Myc plasmid. QPCR and immunohistochemistry experiments were performed to detect the expression of SGLT2 in HCC tissues. In vivo experiments were conducted to corroborate the upregulation of SGLT2 following c-Myc transfection. In invo and vitro investigations were conducted to evaluate the anti-cancer effects of two SGLT2i: EMPA and canagliflozin (CANA). Network pharmacology, molecular docking analyses, CETSA experiments, and additional western blot experiments were used to reveal EMPA's interaction inhibition with mTOR. RESULTS The study identified an increase in SGLT2 expression in HCC tissues as a result of c-Myc overexpression. In vitro experiments confirmed the upregulation of SGLT2 following c-Myc transfection. Notably, the administration of SGLT2i effectively curtailed liver cancer progression, and reduced hepatic fat accumulation in mice. EMPA exhibited significant suppression of cell proliferation in c-Myc-transfected cells. In vitro experiments unveiled EMPA's interaction and with inhibition the activation of mTOR. CONCLUSION Our study highlights EMPA's potential as a therapeutic agent in delaying the development and progression of HCC.
Collapse
Affiliation(s)
- Huiling Rao
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Department of Medical Engineering, The First Affiliated Hospital of Army Medical University, Chongqing 400000, People's Republic of China
| | - Xiaotong An
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Xinyang Qu
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Juan Yu
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Department of Anesthesiology, People's Hospital of Yunxi County of Hubei Province, Yunxi 442600, People's Republic of China
| | - Jin Xie
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Jing Ke
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Zhixin Liu
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Lei You
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lin Tian
- Department of Pathology, Renming Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Weixing Du
- Department of Pathology, Renming Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Wanrong Li
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Jie Jia
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China.
| | - Danwen Liu
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China.
| | - Shan Li
- School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, People's Republic of China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, People's Republic of China.
| |
Collapse
|
2
|
Sun Q, Lei X, Yang X. The crosstalk between non-coding RNAs and oxidative stress in cancer progression. Genes Dis 2025; 12:101286. [PMID: 40028033 PMCID: PMC11870203 DOI: 10.1016/j.gendis.2024.101286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2025] Open
Abstract
As living standards elevate, cancers are appearing in growing numbers among younger individuals globally and these risks escalate with advancing years. One of the reasons is that instability in the cancer genome reduces the effectiveness of conventional drug treatments and chemotherapy, compared with more targeted therapies. Previous research has discovered non-coding RNAs' crucial role in shaping genetic networks involved in cancer cell growth and invasion through their influence on messenger RNA production or protein binding. Additionally, the interaction between non-coding RNAs and oxidative stress, a crucial process in cancer advancement, cannot be overlooked. Essentially, oxidative stress results from the negative effects of radicals within the body and ties directly to cancer gene expression and signaling. Therefore, this review focuses on the mechanism between non-coding RNAs and oxidative stress in cancer progression, which is conducive to finding new cancer treatment strategies.
Collapse
Affiliation(s)
- Qiqi Sun
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
3
|
Xiao S, Miao W, Wang L, Wang L, Tang S, Xu H, Yu Y. Regulation of inflammatory cytokines and activation of PI3K/Akt pathway by Yiqi Jiedu Formula in recurrent Herpes Simplex Keratitis: Experimental and network pharmacology evidence. Virus Res 2025; 355:199561. [PMID: 40120648 PMCID: PMC12001097 DOI: 10.1016/j.virusres.2025.199561] [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: 01/07/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE This study investigates the therapeutic effects of the Yiqi Jiedu (YQJD) formula on Herpes Simplex Keratitis (HSK) induced by herpes simplex virus type 1 (HSV-1) and elucidates its mechanisms of action through experimental and network pharmacology approaches. METHODS Active ingredients of the YQJD formula were identified using UPLC-HRMS. Network pharmacology was employed to predict shared targets between YQJD and HSK, focusing on the PI3K/Akt signaling pathway. Molecular docking was performed to assess the interaction between key ingredients and targets. In vivo, an HSK mouse model was used to evaluate the YQJD formula's impact on corneal lesions and inflammatory factors. In vitro, human corneal epithelial cells (HCECs) were infected with HSV-1 to assess the formula's effect on IL-4 expression. RESULTS UPLC-HRMS identified 34 compounds in YQJD, with Isovitexin and Formononetin exhibiting high oral bioavailability. Network analysis revealed 97 intersecting targets, implicating the PI3K/Akt pathway in YQJD's mechanism. Molecular docking showed strong affinities between IL-4, IL-6, and YQJD compounds. In vivo, YQJD significantly improved corneal lesions and modulated the expression of IL-4, IL-6, and AKT. In vitro, YQJD-containing serum regulated IL-4 expression in HCECs post-HSV-1 infection. CONCLUSION The YQJD formula ameliorates Herpes Simplex Keratitis by regulating inflammatory cytokines and activating the PI3K/Akt pathway, offering a potential therapeutic strategy for HSK.
Collapse
Affiliation(s)
- Shuyu Xiao
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.185 Pu'an Road, Huangpu District, Shuguang, Shanghai 201203, China
| | - Wanhong Miao
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.185 Pu'an Road, Huangpu District, Shuguang, Shanghai 201203, China
| | - Leilei Wang
- Department of Ophthalmology, Shanghai Eye Disease Control Center, Shuguang, 200041, China
| | - Lei Wang
- Department of Ophthalmology, Shanghai Eye Disease Control Center, Shuguang, 200041, China
| | - Sisi Tang
- Department of Ophthalmology, Shanghai Songjiang District Fangta Traditional Chinese Medicine Hospital, Shuguang, 201699, China
| | - Huihui Xu
- Department of Ophthalmology, Shanghai Aier Songchen Eye Hospital, Shuguang, 201699, China
| | - Ying Yu
- Department of Ophthalmology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.185 Pu'an Road, Huangpu District, Shuguang, Shanghai 201203, China.
| |
Collapse
|
4
|
Das S, Halder D, Jeyaprakash RS. Computational-guided approach for identification of PI3K alpha inhibitor in the treatment of hepatocellular carcinoma by virtual screening and water map analysis. J Biomol Struct Dyn 2025; 43:3886-3908. [PMID: 38197431 DOI: 10.1080/07391102.2023.2300131] [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: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most deadly disorders, with a relative survival rate of 36% in the last 5 years. After an extensive literature survey and pathophysiology analysis, PI3Kα was found to be a promising biological target as PIK3CA gene upregulation was observed in HCC, resulting in the loss of apoptosis of cells, which leads to uncontrollable growth and proliferation. Due to superior selectivity and promising therapeutic activity, the PI3K-targeted molecule library was selected, and the ligand preparation was executed. The study mainly focused on e-pharmacophore development, virtual screening and receptor-ligand docking analysis. Then, MMGBSA and ADME prediction analysis was performed with the top 10 molecules; for further analysis of ligand-receptor binding affinity at the catalytic binding site, induced fit docking was performed with the top two molecules. The analysis of quantum chemical stability descriptors, i.e., frontier molecular orbital analysis, was performed followed by molecular dynamics simulation of 100 ns to better understand the ligand-receptor binding. In this study, water map analysis played a significant role in the hit optimization and analysis of the thermodynamic properties of the receptor-ligand complex. The two hit molecules K894-1435 and K894-1045 represented superior docking scores, enhanced stability, and inhibitory action targeting Valine 851 amino acid residue at the catalytic binding site. Hence, the study has significance for the quest for selective PI3Kα inhibitors through the process of hit-to-lead optimization.
Collapse
Affiliation(s)
- Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Debojyoti Halder
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - R S Jeyaprakash
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
5
|
Xi Y, Huang Y, Hu J, Wang Y, Qian Q, Tu L, Nie H, Zhu J, Ding C, Gao X, Zheng X, Huang D, Cheng L. EIF2B5 promotes malignant progression of hepatocellular carcinoma by activating the PI3K/AKT signaling pathway through targeting RPL6. Cell Signal 2025; 132:111821. [PMID: 40246131 DOI: 10.1016/j.cellsig.2025.111821] [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: 11/26/2024] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with limited treatment options and poor prognosis. In this study, we demonstrated the critical role of EIF2B5 in driving HCC progression. We found EIF2B5 expression is significantly upregulated in HCC tumor tissues in several bioinformatics datasets, including The Cancer Genome Atlas, and that high expression of EIF2B5 predicts poor prognosis for HCC patients. Through a series of in vitro cell biology experiments, we found that EIF2B5 knockdown significantly attenuated Hep3B and HepG2 proliferation, migration, and invasion and increased cell cycle arrest, whereas EIF2B5 overexpression promoted HCC progression. Through mass spectrometry and immunoprecipitation validation, we found that EIF2B5 directly interacted with RPL6 and that when EIF2B5 was overexpressed in HCC cells, it promoted the expression of the downstream protein RPL6, which was able to activate the phosphatidylinositol kinase (PI3K)/serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) pathway and thereby increase the proliferation and invasion ability of HCC cell lines, as verified by second-generation sequencing analysis and western blot. We further verified these findings using the mouse ectopic tumor assay, and the results showed that EIF2B5 knockdown significantly inhibited tumor progression in HCC mice. The present study suggests that EIF2B5 promotes malignant progression of HCC by interacting with RPL6 and activating the PI3K/AKT/mTOR signaling pathway and may serve as a potential target for the treatment of HCC.
Collapse
Affiliation(s)
- Yiling Xi
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yue Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiahui Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Wang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiyi Qian
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Linglan Tu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huizong Nie
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiayao Zhu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenguang Ding
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaotao Gao
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoliang Zheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liyan Cheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Wang X, Li J, Nong J, Deng X, Chen Y, Han B, Zeng L, Huang X. MiR-518b Promotes the Tumorigenesis of Hepatocellular Carcinoma by Targeting EGR1 to Regulate PI3K/AKT/mTOR Signaling Pathway. Cell Biochem Biophys 2025:10.1007/s12013-025-01752-z. [PMID: 40221539 DOI: 10.1007/s12013-025-01752-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignancy originating from hepatocytes and is characterized by high invasiveness and fatality. Dysregulation of microRNAs (miRNAs) is frequently observed during HCC progression. This study aimed to investigate the role of miR-518b in HCC cell malignancy and tumor growth. MiR-518b expression in HCC cells was measured by RT-qPCR. The proliferative, migratory and invasive capabilities of Hep3B and SNU-387 were assessed by colony formation, wound healing and transwell assays, respectively. RNA immunoprecipitation and luciferase reporter assays were utilized to verify the binding between miR-518b and its target gene, early growth response factor 1 (EGR1). Results revealed that miR-518b was highly expressed while EGR1 was downregulated in HCC cells. Knockdown of miR-518b significantly repressed cell proliferation, migration and invasion. Moreover, miR-518b bound to 3'untranslated region of EGR1 and negatively regulated its expression in HCC cells. EGR1 knockdown counteracted the inhibitory impact of miR-518b inhibition on malignant cell behaviors. In addition, the silencing of EGR1 activated the PI3K/AKT/mTOR signaling in HCC cells, while miR-518b depletion had the opposite effect. Importantly, the suppressive impact of miR-518b on the pathway was rescued by EGR1 knockdown. In vivo experiments demonstrated that inhibition of miR-518b suppressed HCC tumor growth, reduced EGR1 and Ki67 (a proliferation marker) expression, and inactivated the PI3K/AKT/mTOR signaling. In conclusion, miR-518b promotes HCC tumorigenesis by targeting EGR1 and regulating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xinyuan Wang
- College of Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Juan Li
- Department of pediatrics, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Jiao Nong
- College of Osteopathy, Guangxi University of Chinese Medicine, Nanning, China
| | - Xin Deng
- School of basic medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Yiping Chen
- Emergency department, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Bing Han
- Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Zeng
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xiabing Huang
- Emergency department, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China.
| |
Collapse
|
7
|
Tong W, Qin N, Lu T, Liu L, Liu R, Chen J, Luo N. Integrating bulk and single-cell RNA sequencing reveals SH3D21 promotes hepatocellular carcinoma progression by activating the PI3K/AKT/mTOR pathway. PLoS One 2025; 20:e0302766. [PMID: 40179068 PMCID: PMC11967960 DOI: 10.1371/journal.pone.0302766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/16/2025] [Indexed: 04/05/2025] Open
Abstract
As a novel genetic biomarker, the potential role of SH3D21 in hepatocellular carcinoma remains unclear. Here, we decipher the expression and function of SH3D21 in human hepatocellular carcinoma. The expression level and clinical significance of SH3D21 in hepatocellular carcinoma patients, the relationship between SH3D21 and the features of tumor microenvironment (TME) and role of SH3D21 in promoting hepatocellular carcinoma progression were analyzed based on the bulk samples obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Single-cell sequencing samples from Gene Expression Omnibus (GEO) database were employed to verify the prediction mechanism. Additionally, different biological effects of SH3D21 on hepatocellular carcinoma cells were investigated by qRT-PCR, CCK-8 assay, colony forming assay and Western blot analysis. Bioinformatics analysis and in vitro experiments revealed that the expression level of SH3D21 was up-regulated in hepatocellular carcinoma and correlated with the poor prognosis in hepatocellular carcinoma patients. SH3D21 effectively promoted the proliferation, invasion, and migration as well as the formation of immunosuppressive microenvironment of hepatocellular carcinoma. In addition, SH3D21 can activate the PI3K/AKT/mTOR signaling pathway. SH3D21 stimulates the progression of hepatocellular carcinoma by activating the PI3K/AKT/mTOR signaling pathway, and SH3D21 can serve as a prognostic biomarker and therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wangxia Tong
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Na Qin
- The Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Tao Lu
- Department of hepatobiliary surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Li Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Rong Liu
- Department of Hepatology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jibing Chen
- Centre for Translational Medical Research in Integrative Chinese and Western Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Ning Luo
- Department of Neurology, RuiKang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
8
|
Yang LL, Chen X, Huang KT, Tang ST, Ye GY, Wang JL. BEND3 promotes hepatocellular carcinoma progression and metastasis by activating the PI3K/AKT/mTOR pathway and inducing epithelial-mesenchymal transition. Clin Res Hepatol Gastroenterol 2025; 49:102582. [PMID: 40158797 DOI: 10.1016/j.clinre.2025.102582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE This study aimed to investigate the expression of BEND3 in hepatocellular carcinoma (HCC), its correlation with clinical characteristics, and its functional and mechanistic impacts on HCC progression. METHODS Bioinformatics analyses identified BEND3 as highly expressed in HCC and associated with poor clinical prognosis, which was further validated using qRT-PCR, western blotting and immunohistochemistry. Stable BEND3-overexpressing and silenced cell lines were constructed to evaluate its functional effects. CCK-8 and colony formation assays assessed its influence on cell proliferation, while wound healing and Transwell assays evaluated its role in migration and invasion. WB and immunofluorescence were employed to analyze the effects of BEND3 on epithelial-mesenchymal transition (EMT) and the PI3K/AKT/mTOR signaling pathway. RESULTS Public database analysis, alongside qRT-PCR, western blotting, and immunohistochemical, confirmed that BEND3 expression is significantly elevated in HCC tissues compared to normal liver tissues and is closely associated with poor prognosis. Functional assays demonstrated that BEND3 promotes HCC cell proliferation, migration, and invasion. Mechanistic studies revealed that BEND3 drives HCC progression by inducing EMT and activating the PI3K/AKT/mTOR signaling pathway. CONCLUSION BEND3 is highly expressed in HCC and strongly correlates with poor clinical outcomes. Functional and mechanistic analyses indicate that BEND3 enhances HCC progression by promoting proliferation, migration and invasion via EMT induction and PI3K/AKT/mTOR pathway activation.
Collapse
Affiliation(s)
- Liu-Lin Yang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Xing Chen
- Department of Ultrasonography, Guangxi Medical University First Affiliated Hospital, China
| | - Kai-Ting Huang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Shao-Tong Tang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Gui-Yan Ye
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Ji-Long Wang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China.
| |
Collapse
|
9
|
Gao X, Bian T, Wei X, Xu M, Ren J, Zhao Y, Zheng Y, Li X. Exploration of mechanism of Bufalin and Bufalin derivatives in hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156686. [PMID: 40220402 DOI: 10.1016/j.phymed.2025.156686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Bufalin, a promising candidate for the treatment of hepatocellular carcinoma (HCC), has garnered interest from researchers in exploring its pharmacological mechanisms. The traditional single-target research method makes it difficult to elucidate the molecular mechanism systematically, and there are still limitations. PURPOSE To explore the synergistic mechanisms of Bufalin and its derivatives in the treatment of HCC through quantitative and integrated analysis of big data, this study provides a reference for clinical screening of anti-liver cancer drugs and accelerates the research and development of new drugs. METHODS The literature on the anti-liver cancer activity of Bufalin was searched, the research trends and hotspots were analyzed, and the hot targets were identified. Potential targets and signaling pathways were predicted and compared with established targets. To evaluate the affinity of Bufalin and its derivatives with key targets and validate the results of network pharmacology. The structure-activity relationships and pathways were reviewed, and key pharmacophore and potential pathways were identified. RESULTS Analysis of 302 literatures showed that Na+/K+-ATPase was a hot target in the anti-liver cancer mechanism of Bufalin. 76 potential targets are primarily associated with pathways such as PI3K/AKT/mTOR, Hedgehog, and AMPK/mTOR, which play roles in regulating key targets like phosphoinositide 3-kinase (PIK3), adenosine monophosphate-activated protein kinase (AMPK), and epidermal growth factor receptor (EGFR). Bufalin derivatives have a higher affinity with key targets. The cdocker interaction energy (CIE) of BF211, compound 1, and compound 2 with PI3K were -48.0722 kcal/mol, -50.8791 kcal/mol, and -59.7326 kcal/mol, respectively, which were significantly lower than Bufalin (-26.0859 kcal/mol). CONCLUSIONS Bufalin and its derivatives have significant anti-HCC activity and show good potential for drug development. Their mechanism of action involves a complex network of AMPK targets and PI3K/AKT/mTOR signaling pathways. In the future, specific inhibitors should be developed against these targets and pathways as a new therapeutic strategy for HCC. Among them, compounds 2, 7, and 8 have a higher affinity for potential targets than Bufalin and its other derivatives and have higher research value. In addition, α-pyrone is the key pharmacophore of Bufalin, and structural modification of the C3 position can significantly regulate its cytotoxicity and solubility. Therefore, optimization of the structure of the C3 position is expected to reduce the toxicity and improve the water solubility of Bufalin, overcoming the limitations of its clinical application and providing a safer and more effective solution for the treatment of HCC.
Collapse
Affiliation(s)
- Xinchen Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tianrun Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyu Wei
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mengsi Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Ren
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Zhao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanchao Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xiankuan Li
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
10
|
Zamanian MY, Maleki S, Oghenemaro EF, Singh M, Mohammadi M, Alkhayyat AH, Sapaev IB, Kaur P, Shirsalimi N, Nagarwal A. Omentin-1 as a promising biomarker and therapeutic target in hypertension and heart failure: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04008-y. [PMID: 40126671 DOI: 10.1007/s00210-025-04008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
Omentin-1, a novel adipocytokine predominantly secreted by visceral adipose tissue, has emerged as a significant factor in cardiovascular health, particularly regarding hypertension (HTN) and heart failure (HF). This manuscript investigates the multifaceted roles of omentin-1 in these conditions, emphasizing its protective effects on vascular function and its potential as both a biomarker and therapeutic target. Clinical studies indicate that reduced circulating levels of omentin-1 are associated with metabolic syndrome (MetS) and increased cardiovascular risk, while animal studies demonstrate its ability to ameliorate endothelial dysfunction and lower blood pressure. Omentin-1 exerts its beneficial effects through various signaling pathways, including AMP-activated protein kinase (AMPK) and protein kinase B (Akt), thereby promoting vasodilation, enhancing insulin sensitivity, and mitigating inflammation. In the context of HF, particularly heart failure with preserved ejection fraction (HFpEF), omentin-1 levels exhibit a negative correlation with diastolic dysfunction and inflammatory markers, suggesting its role in cardiac protection. Additionally, the manuscript discusses the implications of omentin-1 in managing obesity-related cardiovascular diseases and its potential utility as a prognostic marker for adverse outcomes in HF patients. Collectively, omentin-1 represents a promising avenue for research in cardiovascular health, with the potential to inform novel therapeutic strategies aimed at improving outcomes in patients with HTN and HF. Further research is necessary to elucidate the details of omentin-1 function and evaluate its potential in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Saba Maleki
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Guilan University of Medical Sciences (GUMS), Rasht, Guilan Province, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mandeep Singh
- Directorate of Sports and Physical Education, University of Jammu, Jammu, India
| | - Maryam Mohammadi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Hussen Alkhayyat
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ibrokhim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan
- Scientific Researcher, University of Tashkent for Applied Sciences, Str. Gavhar 1, 100149, Tashkent, Uzbekistan
- Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Niyousha Shirsalimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Amritesh Nagarwal
- Department of Cardiology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| |
Collapse
|
11
|
Rahimi-Farsi N, Bostanian F, Shahbazi T, Shamsinejad FS, Bolideei M, Mohseni P, Zangooie A, Boustani F, Shoorei H. Novel oncogenes and tumor suppressor genes in Hepatocellular Carcinoma: Carcinogenesis, progression, and therapeutic targets. Gene 2025; 941:149229. [PMID: 39800198 DOI: 10.1016/j.gene.2025.149229] [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: 03/27/2024] [Revised: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Hepatocellular carcinoma (HCC) is the primary malignancy affecting the liver and the leading cause of mortality among individuals with cirrhosis. This complex disease is associated with various risk factors, including environmental, pathological, and genetic influences, which dysregulate gene expression crucial for the cell cycle and cellular/molecular pathways. The disruption of the balance between tumor suppressors and proto-oncogenes amplifies the pathogenic cascade. Given its predilection for diseased or cirrhotic livers and late-stage diagnosis, HCC prognosis is typically poor. Current therapies offer limited benefits, with conventional non-specific cytotoxic agents exhibiting suboptimal efficacy. However, molecularly targeted therapies have emerged as a promising avenue, leveraging the strategic inhibition of carcinogenic molecules to provide heightened specificity and potency compared to cytotoxic chemotherapy. Several clinical trials have demonstrated promising outcomes in advanced HCC with targeted pharmacotherapies. Many genes have been implicated in HCC pathogenesis, underscoring the need to elucidate their molecular functions and roles. This has profound implications for early HCC prognostication via biomarkers and for identifying therapeutic targets to impede neoplastic progression. Notably, evidence highlights the pivotal roles of oncogenes and tumor suppressors in HCC pathophysiology. This discourse examines the potential involvement of ABL1, Annexins, FAK, FOX, and KIF as candidate oncogenes, contrasted with SORBS2, HPCAL1, PCDH10, PLAC8, and CXXC5 as plausible tumor suppressors. Their signaling cascades and relevance to HCC prognosis and progression are delineated to identify targets for improving HCC diagnosis, prognostication, and therapy.
Collapse
Affiliation(s)
| | | | - Taha Shahbazi
- Neurosurgery Research Group (NRG), Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Parvin Mohseni
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Alireza Zangooie
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farnaz Boustani
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran; Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
12
|
Liu D, Pei H, Yao K, Gao J, Chen H, Tong P. Ovalbumin alters DAF-16 Class-II/I gene expressions via insulin/insulin-like growth factor-1 signaling to initiate the innate immune response of Caenorhabditis elegans. Mol Immunol 2025; 179:116-127. [PMID: 39954628 DOI: 10.1016/j.molimm.2025.02.007] [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: 12/19/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Innate immunity, as a significant defense system of the body, plays a key role in allergic reactions, but the mechanism of how food allergens trigger innate immune signaling is still unclear. Ovalbumin (OVA) is a model allergen in food allergy studies. Previous studies by our group have demonstrated that the innate immunity of Caenorhabditis elegans (C. elegans) elicited by OVA treatment was related to the insulin/insulin-like growth factor-1 signaling (IIS) pathway, but the details remain unknown. Therefore, in this study, the molecular mechanism of innate immune signaling transduction of C. elegans stimulated by OVA was determined using genetic mutations as well as RT-PCR, GFP fluorescence visualization monitoring, and slow-killing experiments. Results showed that the expression levels of DAF-16-class-I/II genes in the IIS pathway were significantly changed in C. elegans after OVA treatment, and the upstream gene daf-2 played an important role, which up-regulated the levels of DAF-16-class-II genes dod-22 and F55G11.8 by the daf-2-pqm-1 pathway, and down-regulated the level of DAF-16-class-I gene thn-2 by the daf-2-daf-16 pathway. Moreover, the upstream genes daf-2 and nhr-14, and the transcription factors DAF-16, PQM-1, and SKN-1 in the IIS pathway all participated in the up-regulations of DAF-16-class-II genes dod-17, dod-24, and F55G11.2. In conclusion, details of OVA activating innate immunity in C. elegans through the IIS pathway are reported here, and the results can be further extrapolated to mammals, which will contribute to a better understanding of the mechanism of the occurrence of food allergic reactions from the perspective of innate immunity.
Collapse
Affiliation(s)
- Dong Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; College of Food Science & Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang 330047, PR China
| | - Haibing Pei
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; College of Food Science & Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang 330047, PR China
| | - Kexin Yao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; College of Food Science & Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang 330047, PR China
| | - Jinyan Gao
- College of Food Science & Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang 330047, PR China.
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang 330047, PR China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang 330047, PR China.
| |
Collapse
|
13
|
Kim JS, Choi WM, Kim HI, Chung SW, Choi J, Lee D, Kim KM. Synergistic effects of L-arginine and argininosuccinate synthetase 1 in inducing apoptosis in hepatocellular carcinoma. JOURNAL OF LIVER CANCER 2025; 25:79-90. [PMID: 39806913 PMCID: PMC12010820 DOI: 10.17998/jlc.2024.12.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUNDS/AIMS Hepatocellular carcinoma (HCC) is a malignant cancer with an increasing incidence worldwide. Although numerous efforts have been made to identify effective therapies for HCC, current strategies have limitations. We present a new approach for targeting L-arginine and argininosuccinate synthetase 1 (ASS1). METHODS ASS1 expression in HCC cell lines and primary hepatocytes was detected using polymerase chain reaction and western blotting. Proliferation, migration, signaling pathways, and nitric oxide production in HCC cell lines were measured using MTS, colony formation, wound healing, Western blot, and Griess assays. RESULTS ASS1 expression varied among the HCC cell lines, and cisplatin cytotoxicity was ASS1-dependent. L-arginine alone induced apoptosis in HCC cell lines, regardless of ASS1 expression; however, its effect was enhanced in ASS1-expressing HCC cell lines. Cisplatin cytotoxicity also increased, suggesting that L-arginine acts as a sensitizer to cisplatin in HCC cell lines. ASS1 and L-arginine produced nitric oxide and inhibited key proliferation- and survival-related signaling pathways such as PI3K/Akt and MAPK. Additionally, ASS1 and L-arginine reduced the expression of PKM1 and PKM2 in the glycolysis pathway. CONCLUSIONS Our study revealed that ASS1 and L-arginine exhibited anticancer effects in HCC and sensitized cisplatin-resistant HCC cells to chemotherapy. The combination of ASS1 and L-arginine significantly enhanced the anticancer effects, even in HCC cell lines with low or absent ASS1 expression. These findings highlight the critical roles of arginine and ASS1 in HCC and suggest that increasing arginine availability could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Jin Sun Kim
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Won-Mook Choi
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ha-Il Kim
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Won Chung
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jonggi Choi
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Danbi Lee
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kang Mo Kim
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Ke RS, Dai Y, Tu YL, Liu ZH, Huang KZ, Zhang FX. COLEC10: A potential tumor suppressor and prognostic biomarker in hepatocellular carcinoma through modulation of EMT and PI3K-AKT pathways. Open Life Sci 2025; 20:20220988. [PMID: 40026364 PMCID: PMC11868708 DOI: 10.1515/biol-2022-0988] [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: 06/07/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 03/05/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a cancer with poor prognosis, underscoring the urgent need for enhanced detection and management. This study aimed to investigate the role of Collectin Subfamily Member 10 (COLEC10) in HCC, which was revealed to be associated with various diseases. Bioinformatics tools, including GEO, cBioPortal, and TCGA, were used to identify differentially expressed genes. The prognostic significance of COLEC10 was assessed in two patient cohorts, and its functional impact on Hep3B and SMMC7721 cells was evaluated through CCK-8 and Transwell assays. The underlying mechanisms of COLEC10 in HCC progression were explored using flow cytometry and western blot. COLEC10 was downregulated in HCC and associated with poorer overall survival and disease progression. The potential interaction of COLEC10, CCBE1, and FCN3 was predicted. COLEC10, CCBE1, and FCN3 were identified as prognostic indicators for HCC. Overexpression of COLEC10 inhibited the proliferation, migration, and invasion of HCC cells. COLEC10 overexpression induced G0/G1 cell cycle arrest and suppressed epithelial-mesenchymal transition (EMT), COLEC10 regulated protein expression in the Hedgehog pathway and phosphorylation of key proteins in the PI3K-AKT pathway. COLEC10 is an independent prognostic factor of HCC. COLEC10 regulates EMT, Hedgehog, and PI3K-AKT pathways, providing new ideas for targeted therapy of HCC.
Collapse
Affiliation(s)
- Rui-Sheng Ke
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, Fujian, China
| | - Yun Dai
- Endoscopic Diagnosis and Treatment Department, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
| | - Yan-ling Tu
- Department of Neurology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, 361001, China
| | - Zhao-Hui Liu
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No. 55, Zhenhai Road, Siming District, Xiamen 361003, Fujian, China
| | - Kun-Zhai Huang
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No. 55, Zhenhai Road, Siming District, Xiamen 361003, Fujian, China
| | - Fu-Xing Zhang
- Department of General Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No. 55, Zhenhai Road, Siming District, Xiamen 361003, Fujian, China
| |
Collapse
|
15
|
Li Y, Yin L, Liu B, Liu Y, He D, Liu X, Liu R. Targeting RECQL4 in hepatocellular carcinoma: from prognosis to therapeutic potential. BMC Med Genomics 2025; 18:38. [PMID: 40001159 PMCID: PMC11863661 DOI: 10.1186/s12920-025-02107-6] [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: 11/02/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE The aim of this study is to assess the clinical utility of RecQ Like Helicase 4 (RECQL4) as a prognostic marker in hepatocellular carcinoma (HCC) and investigate its associations with various biological processes, angiogenesis-related factors, immune cell infiltration, immune checkpoints, and drug sensitivity. METHODS RECQL4 expression was analyzed across a range of cancer types utilizing data from the TCGA database. Disparities in RECQL4 expression levels between normal and malignant tissues were evaluated, alongside an analysis of progression-free interval (PFI), disease-specific survival (DSS), and overall survival (OS) curves. Exploration of pertinent pathways, immune cell infiltration, single-cell RNA-seq data, and drug sensitivity was conducted employing The Cancer Genome Atlas (TCGA) and Tumor Immune Single-Cell Hub (TISCH) databases. Furthermore, validation of in-silico results was validated through qPCR, Western blotting, CCK-8 assay, EdU assay, clonogenic assay, wound-healing assay, and transwell assay. RESULTS In HCC, RECQL4 was highly expressed and associated with poorer prognosis (p < 0.05). It positively correlated with pathways related to MYC targets, DNA replication, PI3K/AKT/mTOR signaling, DNA repair mechanisms, and the G2/M checkpoint (R > 0.24, p < 0.001). RECQL4 also showed significant correlations with angiogenesis-related genes, including PTK2 (R > 0.4, p < 0.05), suggesting a potential role in angiogenesis regulation. Immune analysis indicated that RECQL4 was associated with immune cell types such as T helper 2 cells, NK CD56bright cells, and follicular helper T cells, suggesting a positive relationship with their infiltration. High RECQL4 expression was also linked to increased sensitivity to drugs including Sorafenib, 5-Fluorouracil, Cisplatin, and Doxorubicin. Cellular experiments showed that RECQL4 expression at the mRNA and protein levels were significantly higher in HCC cell lines Hep3B and Huh7 compared to the normal liver cell line MHA. Moreover, RECQL4 knockdown resulted in reduced proliferation and migration in HCC cell lines (p < 0.05). CONCLUSIONS RECQL4 shows promise as a biomarker for predicting recurrence and survival in HCC and may affect angiogenesis regulation. Its expression also appears to impact sensitivity to drugs such as Sorafenib, 5-Fluorouracil, Cisplatin, and Doxorubicin. Furthermore, silencing RECQL4 significantly inhibits HCC cell line proliferation and migration.
Collapse
Affiliation(s)
- Yingchen Li
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Linan Yin
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Bowen Liu
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Yan Liu
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Dongfeng He
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Xuesong Liu
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China
| | - Ruibao Liu
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, No. 246 Baojian Road, Harbin, 150086, Heilongjiang Province, China.
| |
Collapse
|
16
|
Wu J, Zhao X, Ren B, Duan X, Sun J. Radioresistance in Hepatocellular Carcinoma: Biological Bases and Therapeutic Implications. Int J Mol Sci 2025; 26:1839. [PMID: 40076465 PMCID: PMC11899467 DOI: 10.3390/ijms26051839] [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: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality. Radiotherapy technology is a common treatment modality that can be used in all stages of HCC. However, in some cases, radiotherapy fails in clinical practice mainly because of the patient's resistance to radiotherapy, creating a bottleneck for future breakthroughs. HCC radiosensitivity is primarily related to DNA double-strand break repair, cellular autophagy, cell cycle, cellular metabolism, and hypoxic environmental regulators. Therefore, a comprehensive understanding of its molecular mechanisms will be of immense importance in reversing HCC radioresistance. In this review, we provide a comprehensive overview of the mechanism of action of radiotherapy on HCC, the cellular and molecular basis of radiation resistance in HCC, related treatment modalities, and future prospects.
Collapse
Affiliation(s)
- Jianhui Wu
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| | - Xiaofang Zhao
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| | - Bowen Ren
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| | - Xuezhang Duan
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
| | - Jing Sun
- Department of Radiation Oncology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China; (J.W.); (X.Z.)
- Medical School, Chinese PLA General Hospital, Beijing 100853, China;
| |
Collapse
|
17
|
Huo Y, Ma M, Tian W, Wang F, Liao X. Hepatic adverse events associated with anaplastic lymphoma kinase tyrosine kinase inhibitors: a disproportionality analysis based on FAERS database and analysis of drug-gene interaction network. Expert Opin Drug Saf 2025:1-11. [PMID: 39949054 DOI: 10.1080/14740338.2025.2467830] [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: 09/27/2024] [Revised: 01/06/2025] [Accepted: 01/24/2025] [Indexed: 02/19/2025]
Abstract
BACKGROUND Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) are vital for treating ALK-positive cancers but have been associated with liver injury, necessitating further safety investigation. This study examines hepatic adverse event (AE) signals related to ALK TKIs using the U.S. FDA Adverse Event Reporting System (FAERS) and explores potential mechanisms of liver injury. RESEARCH DESIGN AND METHODS AE reports from FAERS (Q3 2011 to Q1 2024) related to liver injury were analyzed using the reporting odds ratio (ROR) and multi-item gamma Poisson shrinker (MGPS) methods. Pathway enrichment and drug-gene network analyses were performed to investigate underlying mechanisms. RESULTS This study identified 2,132 AE reports from the FAERS database linking hepatic AEs to ALK TKIs therapy. Significant signals were detected by ROR and MGPS methods, with common AEs including aminotransferase abnormalities, hyperbilirubinemia, and increased blood alkaline phosphatase, mainly occurring within the first 30 days of treatment. Gene analysis revealed key nodes in the protein-protein interaction (PPI) network, such as PIK3CA, SRC, and PTK2. Enriched KEGG pathways included the MAPK, PI3K-Akt, and Ras signaling. CONCLUSION This pharmacovigilance study identifies significant AE signals linking ALK TKIs to liver injury, highlighting potential mechanisms and providing insights for clinical management and patient outcomes.
Collapse
Affiliation(s)
- Yan Huo
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Minghua Ma
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weiwei Tian
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fang Wang
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaolan Liao
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
18
|
Zheng J, Wang S, Xia L, Sun Z, Chan KM, Bernards R, Qin W, Chen J, Xia Q, Jin H. Hepatocellular carcinoma: signaling pathways and therapeutic advances. Signal Transduct Target Ther 2025; 10:35. [PMID: 39915447 PMCID: PMC11802921 DOI: 10.1038/s41392-024-02075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 02/09/2025] Open
Abstract
Liver cancer represents a major global health concern, with projections indicating that the number of new cases could surpass 1 million annually by 2025. Hepatocellular carcinoma (HCC) constitutes around 90% of liver cancer cases and is primarily linked to factors incluidng aflatoxin, hepatitis B (HBV) and C (HCV), and metabolic disorders. There are no obvious symptoms in the early stage of HCC, which often leads to delays in diagnosis. Therefore, HCC patients usually present with tumors in advanced and incurable stages. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of HCC. Beyond the frequently altered and therapeutically targeted receptor tyrosine kinase (RTK) pathways in HCC, pathways involved in cell differentiation, telomere regulation, epigenetic modification and stress response also provide therapeutic potential. Investigating the key signaling pathways and their inhibitors is pivotal for achieving therapeutic advancements in the management of HCC. At present, the primary therapeutic approaches for advanced HCC are tyrosine kinase inhibitors (TKI), immune checkpoint inhibitors (ICI), and combination regimens. New trials are investigating combination therapies involving ICIs and TKIs or anti-VEGF (endothelial growth factor) therapies, as well as combinations of two immunotherapy regimens. The outcomes of these trials are expected to revolutionize HCC management across all stages. Here, we provide here a comprehensive review of cellular signaling pathways, their therapeutic potential, evidence derived from late-stage clinical trials in HCC and discuss the concepts underlying earlier clinical trials, biomarker identification, and the development of more effective therapeutics for HCC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhen Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, PR China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
19
|
Li L, Li G, Zhai W. Single-cell transcriptomic analysis reveals efferocytosis signature predicting immunotherapy response in hepatocellular carcinoma. Dig Liver Dis 2025:S1590-8658(25)00207-5. [PMID: 39904693 DOI: 10.1016/j.dld.2025.01.196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/25/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a substantial global health challenge owing to its high mortality rate and limited therapeutic options. We aimed to develop an efferocytosis-related gene signature (ER.Sig) and conduct a transcriptomic analysis to predict the prognosis and immunotherapeutic responses of patients with HCC. METHODS Single-cell RNA sequencing data and bulk RNA sequencing data were obtained from public databases. Based on single-sample gene set enrichment analysis and Weighted Gene Co-expression Network analyses, efferocytosis-related genes (ERGs) were selected at both the single-cell and bulk transcriptome levels. A machine-learning framework employing ten different algorithms was used to develop the ER.Sig. Subsequently, a multi-omics approach (encompassing genomic analysis, single-cell transcriptomics, and bulk transcriptomics) was employed to thoroughly elucidate the prognostic signatures. RESULTS Analysis of the HCC single-cell transcriptomes revealed significant efferocytotic activity in macrophages, endothelial cells, and fibroblasts within the HCC microenvironment. We then constructed a weighted co-expression network and identified six modules, among which the brown module (168 genes) was most highly correlated with the efferocytosis score (cor = 0.84). Using the univariate Cox regression analysis, 33 prognostic ERGs were identified. Subsequently, a predictive model was constructed using 10 machine-learning algorithms, with the random survival forest model showing the highest predictive performance. The final model, ER.Sig, comprised nine genes and demonstrated robust prognostic capabilities across multiple datasets. High-risk patients exhibited greater intratumoral heterogeneity and higher TP53 mutation frequencies than did low-risk patients. Immune landscape analysis revealed that compared with high-risk patients, low-risk patients exhibited a more favorable immune environment, characterized by higher proportions of CD8+ T and B cells, tumor microenvironment score, immunophenoscore, and lower Tumor Immune Dysfunction and Exclusion scores, indicating better responses to immunotherapy. Additionally, an examination of an independent immunotherapy cohort (IMvigor210) demonstrated that low-risk patients exhibited more favorable responses to immunotherapy and improved prognoses than did their high-risk counterparts. CONCLUSIONS The developed ER.Sig effectively predicted the prognosis of patients with HCC and revealed significant differences in tumor biology and treatment responses between the risk groups.
Collapse
Affiliation(s)
- Longhu Li
- Department of Intervention, Linfen Central Hospital, Linfen, PR China.
| | - Guangyao Li
- Department of Intervention, Linfen Central Hospital, Linfen, PR China
| | - Wangfeng Zhai
- Department of Intervention, Linfen Central Hospital, Linfen, PR China
| |
Collapse
|
20
|
Li J, Xue J, Liu T, Feng Y, Xu N, Huang J, Yin Y, Zhang J, Mou H, Shentu J, Bao H, Xu Z, Xu Z. Phase Ib study of the oral PI3Kδ inhibitor linperlisib in patients with advanced solid tumors. Int J Clin Oncol 2025; 30:241-251. [PMID: 39538003 PMCID: PMC11785675 DOI: 10.1007/s10147-024-02657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Patients with advanced solid tumors have a suboptimal prognosis. This study investigated the safety and feasibility of linperlisib, a selective phosphatidylinositol 3-kinase delta isoform (PI3Kδ) inhibitor, for treating patients with advanced solid tumors. METHODS In this phase Ib, single-arm, open-label, multi-center clinical trial, patients with histologically confirmed advanced solid tumors from eight centers in China were enrolled to receive oral linperlisib (80 mg/day). The primary endpoint was safety. RESULTS Between August 2019 and June 2022, 94 patients were enrolled in the trial and received the study treatment. The most common (≥ 20%) treatment emergent adverse events (TEAEs) of all grades irrespective of causality were increased aspartate aminotransferase (AST) (26.6%), proteinuria (26.6%), decreased appetite (25.5%), increased alanine aminotransferase (ALT) (22.3%), weight loss (21.3%), and anemia (21.3%). The most common grade ≥ 3 TEAEs were diarrhea (4.3%), increased AST (3.2%), increased ALT (3.2%), neutropenia (3.2%), anemia (3.2%), increased blood alkaline phosphatase (3.2%). The objective response rate (ORR) was 1.1% (95% confidence interval [CI] 0.0-5.8), and the disease control rate (DCR) was 37.2% (95% CI 27.5-47.8). As of the data cutoff, the median follow-up time was 4.2 months (95% CI 2.8-6.9). The median progression-free survival (PFS) was 1.85 months (95% CI 1.79-1.88). The median overall survival (OS) was not reached. CONCLUSION Linperlisib showed an acceptable safety profile and preliminary clinical benefit in patients with a range of advanced solid tumors. Further studies of linperlisib safety and efficacy are warranted.
Collapse
Affiliation(s)
- Jin Li
- Department of Oncology, East Hospital Affiliated to Tongji University, No. 150 Jimo Road, Pudong New Area, Shanghai, 200120, China.
| | - Junli Xue
- Department of Oncology, East Hospital Affiliated to Tongji University, No. 150 Jimo Road, Pudong New Area, Shanghai, 200120, China
| | - Tianshu Liu
- Department of Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032, China
| | - Yi Feng
- Department of Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032, China
| | - Nong Xu
- Department of Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Jianjin Huang
- Department of Oncology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Yongmei Yin
- Department of Oncology, Jiangsu Provincial People's Hospital, Nanjing, 210029, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haibo Mou
- Department of Oncology, Zhejiang Shulan Hospital, Hangzhou, 310022, China
| | - Jiangzhong Shentu
- Department of Pharmacology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Hanying Bao
- Shanghai Yingli Pharmaceutical Co., Ltd., Shanghai, 201210, China
| | - Zusheng Xu
- Shanghai Yingli Pharmaceutical Co., Ltd., Shanghai, 201210, China
| | - Zuhong Xu
- Shanghai Yingli Pharmaceutical Co., Ltd., Shanghai, 201210, China
| |
Collapse
|
21
|
Wang F, Han X, Han Z, Wang J, Cai Z, Chen G, Bai D, Cui W. Slide-Ring Structured Stress-Electric Coupling Hydrogel Microspheres for Low-Loss Transduction Between Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413156. [PMID: 39744759 DOI: 10.1002/adma.202413156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/19/2024] [Indexed: 02/20/2025]
Abstract
High transductive loss at tissue injury sites impedes repair. The high dissipation characteristics in the electromechanical conversion of piezoelectric biomaterials pose a challenge. Therefore, supramolecular engineering and microfluidic technology is utilized to introduce slide-ring polyrotaxane and conductive polypyrrole to construct stress-electric coupling hydrogel microspheres. The molecular slippage mechanism of slide-ring structure stores and releases mechanical energy, reducing mechanical loss, the piezoelectric barium titanate enables stress-electricity conversion, and conjugated π-electron movement in conductive network improves the internal electron transfer efficiency of microspheres, thereby reducing the loss in stress-electricity conversion for the first time. Compared to traditional piezoelectric hydrogel microspheres, the stress-electric coupling efficiency of low-dissipation microspheres increased by 2.3 times, and the energy dissipation decreased to 43%. At cellular level, electrical signals generated by the microspheres triggered Ca2+ influx into stem cells and upregulated the cAMP signaling pathways, promoting chondrogenic differentiation. Enhanced electrical signals induced macrophage polarization to the M2 phenotype, reshaping inflammation and promoting tissue repair. In vivo, the low-dissipation microspheres restored low-loss transduction between tissues, alleviated cartilage damage, improved behavioral outcomes, and promoted the treatment of osteoarthritis in rats. Therefore, this study proposes a new strategy for restoring low-loss transduction between tissues, particularly in mechanically sensitive tissues.
Collapse
Affiliation(s)
- Fan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, the Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, P. R. China
| | - Xiaoyu Han
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Zeyu Han
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Juan Wang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Zhengwei Cai
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Gang Chen
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, the Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, P. R. China
| | - Dingqun Bai
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
22
|
Dong L, Dong C, Yu Y, Jiao X, Zhang X, Zhang X, Li Z. Transcriptomic analysis of Paraoxonase 1 expression in hepatocellular carcinoma and its potential impact on tumor immunity. Clin Transl Oncol 2025; 27:612-629. [PMID: 39031295 DOI: 10.1007/s12094-024-03598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is characterized by a complex pathogenesis that confers aggressive malignancy, leading to a lack of dependable biomarkers for predicting invasion and metastasis, which results in poor prognoses in patients with HCC. Glycogen storage disease (GSD) is an uncommon metabolic disorder marked by hepatomegaly and liver fibrosis. Notably, hepatic adenomas in GSD patients present a heightened risk of malignancy compared to those in individuals without the disorder. In this investigation, PON1 emerged as a potential pivotal gene for HCC through bioinformatics analysis. METHODS Transcriptomic profiling data of liver cancer were collected and integrated from TCGA and GEO databases. Bioinformatics analysis was conducted to identify mutated mRNAs associated with GSD, and the PON1 gene was selected as a key gene. Patients were grouped based on the expression levels of PON1, and differences in clinical characteristics, biological pathways, immune infiltration, and expression of immune checkpoints were compared. RESULTS The expression levels of the PON1 gene showed significant differences between the high-expression group and the low-expression group in HCC patients. Further analysis indicated that the PON1 gene at different expression levels might influence the clinical manifestations, biological processes, immune infiltration, and expression of immune checkpoints in HCC. Additionally, immunohistochemistry (IHC) results revealed high expression of PON1 in normal tissues and low expression in HCC tissues. These findings provide important clues and future research directions for the early diagnosis, prognosis, immunotherapy, and potential molecular interactions of HCC. CONCLUSION Our investigation underscores the noteworthy prognostic significance of PON1 in HCC, suggesting its potential pivotal role in modulating tumor progression and immune cell infiltration. These findings establish PON1 as a novel tumor biomarker with significant implications for the prognosis, targeted therapy, and immunotherapy of patients with HCC.
Collapse
Affiliation(s)
- Linhuan Dong
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Changjun Dong
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Yunlin Yu
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xin Jiao
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xiangwei Zhang
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xianlin Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China.
| | - Zheng Li
- Department of General surgery, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China.
| |
Collapse
|
23
|
Zhang Y, Zhou YL, Xu N, Meng T, Wang ZZ, Pan FM, Zhu LX. Chemokines and PI3K/AKT signaling pathway mediate the spontaneously ruptured hepatocellular carcinoma through the regulation of the cell cycle. Hepatobiliary Pancreat Dis Int 2025:S1499-3872(25)00029-3. [PMID: 39952875 DOI: 10.1016/j.hbpd.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/26/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND The incidence of spontaneously ruptured hepatocellular carcinoma (srHCC) has been shown to significantly elevate mortality rates. However, the precise mechanisms underlying srHCC remain poorly understood. METHODS Analysis was conducted on the data of 198 hepatocellular carcinoma (HCC) patients to investigate the factors contributing to srHCC. The clinical data of 33 transcriptome HCC patients were served for verification. An in-depth transcriptome analysis was conducted to investigate the distinctions between 26 cases of srHCC and 35 cases of non-ruptured hepatocellular carcinoma (nrHCC). Weighted Gene Co-expression Network Analysis (WGCNA) tool was utilized to develop a gene co-expression network. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathways enrichment, and protein-protein interaction (PPI) network were carried out. The corresponding samples for spontaneously ruptured hepatocellular carcinoma tissue (srHCC-T) and ruptured hepatocellular carcinoma paracancerous tissue (srHCC-P) was selected for verification. Transcriptional data were validated through reverse transcription quantitative polymerase chain reaction (RT-qPCR). Immunofluorescence (IF), immunohistochemistry (IHC) and Western blot were used to detect the protein expression. RESULTS Our results showed that white blood cell (WBC) and monocyte levels were significant independent risk factors for srHCC (P < 0.05). There was a strong association between the srHCC-T and the expression of cell cycle-related genes BUB1B and macrophage function-related gene MACRO. Furthermore, chemokines and the PI3K/AKT signaling pathway play a crucial role in regulating the cell cycle process through a complex network of interactions, ultimately impacting the occurrence of srHCC. CONCLUSIONS Our study confirms that chemokines and the PI3K/AKT signaling pathway mediate the occurrence of HCC rupture by regulating the cell cycle. We provide a theoretical basis for the clinical treatment of srHCC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of General Surgery, the Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Yang-Liu Zhou
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Na Xu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Tao Meng
- Department of General Surgery, Hefei First People's Hospital, Hefei 230000, China
| | - Zhen-Zhen Wang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Fa-Ming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei 230032, China.
| | - Li-Xin Zhu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
24
|
Chen C, Wang M, Tu D, Cao J, Zhang C, Bai D. Roles of anoikis in hepatocellular carcinoma: mechanisms and therapeutic potential. Med Oncol 2025; 42:58. [PMID: 39885089 DOI: 10.1007/s12032-025-02612-9] [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: 11/26/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, is a highly aggressive malignancy with limited viable therapeutic options. For early HCC, resection surgery is currently the most effective treatment. However, in advanced stages, resection alone does not sufficiently address the disease, so finding a method with a better prognosis is necessary. Anoikis, known as matrix detachment-induced apoptosis or detachment-induced cell death, is crucial for tissue development and homeostasis. Cancer cells develop means to evade anoikis, e.g. anoikis resistance, thereby allowing for cells to survive under anchorage-independent conditions. HCC cells often acquire resistance to anoikis, allowing them to survive after detaching from the extracellular matrix and contributing to tumor spread. This review discusses the mechanisms of anoikis in HCC, exploring the potential of drug-induced anoikis and targeting anoikis resistance as promising therapeutic strategies for treating HCC, analyzing the value of anoikis in the immune of HCC, and propose potential pathways in oncotherapy, which can provide background knowledge for subsequent related research.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Mengyao Wang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Daoyuan Tu
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Jun Cao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, China.
| |
Collapse
|
25
|
Saraswat I, Goel A, Gupta J. An In-depth Review on Argemone mexicana in the Management of Liver Health and Liver Cancer. Anticancer Agents Med Chem 2025; 25:24-34. [PMID: 39225208 DOI: 10.2174/0118715206307964240821051756] [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: 03/11/2024] [Revised: 05/25/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Argemone mexicana, commonly known as the Mexican prickly poppy, has been historically employed in traditional medicine for various ailments, including liver disorders. Given the rising prevalence of liver diseases, including cancer, investigating the potential efficacy of Argemone mexicana in promoting liver health is of paramount importance. This review aims to provide a comprehensive analysis of the existing literature on the hepatoprotective and anticancer properties of Argemone mexicana. METHODOLOGY A systematic literature search was conducted across PubMed, Google Scholar, and relevant botanical and pharmacological databases. Studies from various sources, including in vitro experiments, animal models, and clinical trials, were included in the review. The search focused on articles published up to 2010-2023, encompassing research that explored the botanical characteristics, chemical composition, traditional uses, and pharmacological properties of Argemone mexicana, specifically emphasizing its impact on liver health and cancer. RESULTS The review revealed a wealth of studies highlighting the diverse pharmacological properties of Argemone mexicana. The botanical composition includes compounds with antioxidant and anti-inflammatory potential, suggesting hepatoprotective effects. Studies using in vitro and in vivo models demonstrated promising outcomes regarding liver function improvement and inhibition of liver cancer cell proliferation. While some clinical studies supported the traditional uses of Argemone mexicana, further well-designed trials are warranted to establish its clinical efficacy. CONCLUSION In conclusion, Argemone mexicana shows promise as a natural agent for promoting liver health and combating liver cancer. Bioactive compounds with antioxidant and anti-inflammatory properties suggest potential hepatoprotective effects. However, translating these findings into clinical practice requires further rigorous investigation, including well-designed clinical trials. This review provides a foundation for future research efforts aimed at elucidating the full therapeutic potential of Argemone mexicana in liver health and cancer management.
Collapse
Affiliation(s)
- Istuti Saraswat
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| | - Anjana Goel
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| | - Jyoti Gupta
- Department of Biotechnology, GLA University, 17km Stone, NH-2 Mathura-Delhi Road Mathura, Chaumuhan, Mathura, Uttar Pradesh, India
| |
Collapse
|
26
|
Chen G, Zhang Y, Zhou Y, Luo H, Guan H, An B. Targeting the mTOR Pathway in Hepatocellular Carcinoma: The Therapeutic Potential of Natural Products. J Inflamm Res 2024; 17:10421-10440. [PMID: 39659752 PMCID: PMC11630751 DOI: 10.2147/jir.s501270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/24/2024] [Indexed: 12/12/2024] Open
Abstract
Despite advancements in cancer treatment through surgery and drugs, hepatocellular carcinoma (HCC) remains a significant challenge, as reflected by its low survival rates. The mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating the cell cycle, proliferation, apoptosis, and metabolism. Notably, dysregulation leading to the activation of the mTOR signaling pathway is common in HCC, making it a key focus for in-depth research and a target for current therapeutic strategies. This review focuses on the role of the mTOR signaling pathway and its downstream effectors in regulating HCC cell proliferation, apoptosis, autophagy, cell cycle, and metabolic reprogramming. Moreover, it emphasizes the potential of natural products as modulators of the mTOR signaling pathway. When incorporated into combination therapies, these natural products have been demonstrated to augment therapeutic efficacy and surmount drug resistance. These products target key signaling pathways such as mTOR signaling pathways. Examples include 11-epi-sinulariolide acetate, matrine, and asparagus polysaccharide. Their inhibitory effects on these processes suggest valuable directions for the development of more effective HCC therapeutic strategies. Various natural products have demonstrated the ability to inhibit mTOR signaling pathway and suppress HCC progression. These phytochemicals, functioning as mTOR signaling pathway inhibitors, hold great promise as potential anti-HCC agents, especially in the context of overcoming chemoresistance and enhancing the outcomes of combination therapies.
Collapse
Affiliation(s)
- Guo Chen
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Ya Zhang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Yaqiao Zhou
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Hao Luo
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Hongzhi Guan
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Baiping An
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
27
|
Rahdan F, Abedi F, Dianat-Moghadam H, Sani MZ, Taghizadeh M, Alizadeh E. Autophagy-based therapy for hepatocellular carcinoma: from standard treatments to combination therapy, oncolytic virotherapy, and targeted nanomedicines. Clin Exp Med 2024; 25:13. [PMID: 39621122 PMCID: PMC11611955 DOI: 10.1007/s10238-024-01527-5] [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: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Human hepatocellular carcinoma (HCC) has been identified as a significant cause of mortality worldwide. In recent years, extensive research has been conducted to understand the underlying mechanisms of autophagy in the pathogenesis of the disease, with the aim of developing novel therapeutic agents. Targeting autophagy with conventional therapies in invasive HCC has opened up new opportunities for treatment. However, the emergence of resistance and the immunosuppressive tumor environment highlight the need for combination therapy or specific targeting, as well as an efficient drug delivery system to ensure targeted tumor areas receive sufficient doses without affecting normal cells or tissues. In this review, we discuss the findings of several studies that have explored autophagy as a potential therapeutic approach in HCC. We also outline the potential and limitations of standard therapies for autophagy modulation in HCC treatment. Additionally, we discuss how different combination therapies, nano-targeted strategies, and oncolytic virotherapy could enhance autophagy-based HCC treatment in future research.
Collapse
Affiliation(s)
- Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Abedi
- Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
| | - Maryam Zamani Sani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Zheng P, Xu D, Cai Y, Zhu L, Xiao Q, Peng W, Chen B. A multi-omic analysis reveals that Gamabufotalin exerts anti-hepatocellular carcinoma effects by regulating amino acid metabolism through targeting STAMBPL1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156094. [PMID: 39348778 DOI: 10.1016/j.phymed.2024.156094] [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: 05/28/2024] [Revised: 09/03/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), a prevalent type of liver cancer, is characterized by an unfavorable prognosis and a high mortality rate. Identifying novel treatments to prevent HCC recurrence and metastasis remains crucial for improving patient survival. Gamabufotalin (CS-6), a primary bufadienolide derived from the traditional Chinese medicine Chansu, has demonstrated significant anti-tumor activity. However, the effects and underlying mechanisms of CS-6 on HCC cells are not yet fully understood. PURPOSE This study sought to elucidate the anti-HCC effects and potential mechanisms of CS-6. In vitro experiments were conducted using the HCC cell lines MHCC97H and Huh-7, employing CCK-8 assays, colony formation assays, wound healing assays, transwell invasion and migration assays, and flow cytometry to assess apoptosis and cell cycle dynamics. A multi-omics approach, including metabolomics and RNA sequencing analysis, was utilized to identify CS-6's molecular targets and mechanisms in HCC therapy. Additionally, in vivo assessments were performed using xenografts in nude mice. RESULTS CS-6 significantly inhibited HCC cell proliferation, migration, and invasion. Multi-omics analysis suggested that CS-6's anti-HCC effects may involve the modulation of metabolic pathways, potentially through the downregulation of STAMBPL1, resulting in reduced mTOR signaling, increased apoptosis, and suppression of malignant HCC behavior. In vivo studies further confirmed that CS-6 significantly suppressed tumor growth and enhanced apoptosis and autophagy within tumors. CONCLUSION These results underscore the therapeutic potential of CS-6 in HCC treatment. The study offers novel insights into the mechanism of CS-6, suggesting that its therapeutic efficacy may be uniquely mediated by targeting STAMBPL1. This distinct mechanism sets CS-6 apart from existing HCC treatments and positions it as a promising candidate for further clinical investigation.
Collapse
Affiliation(s)
- Piao Zheng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Die Xu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yisi Cai
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lemei Zhu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Qiao Xiao
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Academician Workstation, Changsha Medical University, Changsha 410219, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Bolin Chen
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/the affiliated Cancer Hospital of Xiangya school of Medicine, Central South University, No.283 Tongzipo Road, Changsha 410013, China.
| |
Collapse
|
29
|
Bai Z, Li H, Jiao B. Potential Therapeutic Effect of Sinigrin on Diethylnitrosamine-Induced Liver Cancer in Mice: Exploring the Involvement of Nrf-2/HO-1, PI3K-Akt-mTOR Signaling Pathways, and Apoptosis. ACS OMEGA 2024; 9:46064-46073. [PMID: 39583716 PMCID: PMC11579720 DOI: 10.1021/acsomega.4c06203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Sinigrin is a glucosinolate present in plants of the family Brassicaceae and has been considered for its anticancer potential. This study examines the efficacy of sinigrin on the liver cancer caused by diethylnitrosamine (DEN) in mice through the analysis of its impact on the Nrf-2/HO-1, PI3K-Akt-mTOR, and apoptotic pathways. Development of liver cancer was induced by intraperitoneal injection at the age of 14 days with DEN (25 mg/kg) in mice. Thereafter, sinigrin was orally administered at doses of 10 and 20 mg/kg body weight per day the last 28 days. At the end of 10 weeks, mice were sacrificed and then we conducted hepatic biochemical and molecular assessments. Sinigrin reduced the serum level of alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), alpha-fetoprotein (AFP), and bilirubin but increased total protein, and albumin, levels. Sinigrin increased the antioxidant enzymes (SOD, CAT, GPx, and GST) as indicated by reduced 8-OHdG, TBARS and increased glutathione. Sinigrin reduced the levels of inflammatory cytokines (IL-6, IL-1β, TNF-α, and NF-κB p65) and PI3K/AKT/mTOR signaling pathway. Sinigrin also activated the intrinsic mitochondrial apoptosis pathway mediated by p53, downregulated antiapoptotic proteins (Bcl-2), up-regulated pro-apoptosis regulatory proteins like Bax and caspase-3. All these results indicate that the protective effects of sinigrin against liver cancer are likely to be applied as an effective therapeutic agent through its antioxidant and pro-apoptotic activities.
Collapse
Affiliation(s)
- Zhe Bai
- Department
of Hepatobiliary Pancreatic and Gastrosurgery, Shanxi Province Cancer
Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital
Affiliated to Shanxi Medical University, Xinghualing District Workers New Street 3, Taiyuan 030013, China
| | - Hui Li
- Department
of Gastroenterology, The First Hospital
of Shanxi Medical University, No. 85, Jiefang South Road, Taiyuan, Shanxi 030001, China
| | - Baoping Jiao
- Department
of Hepatobiliary Pancreatic and Gastrosurgery, Shanxi Province Cancer
Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital
Affiliated to Shanxi Medical University, Xinghualing District Workers New Street 3, Taiyuan 030013, China
| |
Collapse
|
30
|
Stols-Gonçalves D, Meijnikman AS, Tristão LS, dos Santos CL, Denswil NP, Verheij J, Bernardo WM, Nieuwdorp M. Metabolic Dysfunction-Associated Steatotic Liver Disease and Alcohol-Associated Liver Disease: Liver DNA Methylation Analysis-A Systematic Review. Cells 2024; 13:1893. [PMID: 39594641 PMCID: PMC11592595 DOI: 10.3390/cells13221893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated liver disease (MASLD) and alcohol-associated liver disease (ALD) are among the leading causes of liver disease worldwide. The exact roles of epigenetic factors in both diseases remains largely unknown. In this context, liver DNA methylation remains a field that requires further exploration and understanding. METHODS We performed a systematic review of liver DNA methylation in humans with MASLD or ALD using Ovid MEDLINE, Ovid Embase, and Cochrane Library. We included human studies where liver DNA methylation was assessed in patients with MASLD and/or ALD. The Rayyan platform was used to select studies. Risk of bias was assessed with the "risk of bias in non-randomized studies of interventions" tool, ROBINS-I. We performed pathway analysis using the most important differentially methylated genes selected in each article. RESULTS Fifteen articles were included in this systematic review. The risk of bias was moderate to serious in all articles and bias due to confounding and patient selection was high. Sixteen common pathways, containing differentially methylated genes, including cancer pathways, were identified in both diseases. CONCLUSIONS There are common pathways, containing differentially methylated genes, in ALD and MASLD, such as pathways in cancer and peroxisome proliferator-activated receptor (PPAR) signaling pathways. In MASLD, the insulin signaling pathway is one of the most important, and in ALD, the MAPK signaling pathway is the most important. Our study adds one more piece to the puzzle of the mechanisms involved in steatotic liver disease.
Collapse
Affiliation(s)
- Daniela Stols-Gonçalves
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Meibergdreef 9 (Room A01-112), 1105 AZ Amsterdam, The Netherlands; (A.S.M.); (M.N.)
| | - Abraham S. Meijnikman
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Meibergdreef 9 (Room A01-112), 1105 AZ Amsterdam, The Netherlands; (A.S.M.); (M.N.)
| | - Luca Schiliró Tristão
- Department of Evidence-Based Medicine, Faculdade de Ciências Médicas de Santos—Lusiada University Center, Santos 11050-071, SP, Brazil; (L.S.T.); (C.L.d.S.); (W.M.B.)
| | - Clara Lucato dos Santos
- Department of Evidence-Based Medicine, Faculdade de Ciências Médicas de Santos—Lusiada University Center, Santos 11050-071, SP, Brazil; (L.S.T.); (C.L.d.S.); (W.M.B.)
| | - Nerissa P. Denswil
- Medical Library, Amsterdam University Medical Centre, University of Amsterdam, 1012 WP Amsterdam, The Netherlands;
| | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Centre, 1105 AZ Amsterdam, The Netherlands;
| | - Wanderley M. Bernardo
- Department of Evidence-Based Medicine, Faculdade de Ciências Médicas de Santos—Lusiada University Center, Santos 11050-071, SP, Brazil; (L.S.T.); (C.L.d.S.); (W.M.B.)
- Faculdade de Medicina d Universidade de São Paulo, São Paulo 05508-220, SP, Brazil
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centre, Meibergdreef 9 (Room A01-112), 1105 AZ Amsterdam, The Netherlands; (A.S.M.); (M.N.)
| |
Collapse
|
31
|
Das D, Xie L, Qiao D, Jia J, Hong J. Discovery of novel, orally bioavailable phenylacetamide derivatives as multikinase inhibitors and in vivo efficacy study in hepatocellular carcinoma animal models. Bioorg Med Chem Lett 2024; 113:129971. [PMID: 39306048 DOI: 10.1016/j.bmcl.2024.129971] [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: 07/18/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
Hepatocellular carcinoma (HCC) is considered as one of the leading causes of death in liver disease patients. Several signal transduction pathways are involved in HCC pathogenesis. Multikinase inhibitors (MKIs) show beneficial effects for HCC and the FDA approved a few MKIs including sorafenib, lenvatinib for HCC treatments. Here, a novel series of phenylacetamide derivatives were designed, synthesized and evaluated as multikinase inhibitors. Several compounds showed nanomolar IC50 values against FLT1, FLT3, FLT4, KDR, PDGFRα, PDGFRβ. The compounds were tested against human hepatocellular carcinoma (HCC), human colon adenocarcinoma and human gastric carcinoma cell lines. With favorable pharmacokinetics profiles, compound 12 and compound 14 were selected for in vivo efficacy studies in Hep3B mice models and demonstrated efficacious than sorafenib.
Collapse
Affiliation(s)
- Debasis Das
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China.
| | - Lingzhi Xie
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China
| | - Dandan Qiao
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China
| | - Jianhe Jia
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China
| | - Jian Hong
- Arromax Pharmatech Co. Ltd. Sangtiandao Innovation Park, No. 1 Huayun Road, SIP, Suzhou 215123, PR China.
| |
Collapse
|
32
|
Kang Y, Xia Q, Jiang Y, Li Z. MVGNet: Prediction of PI3K Inhibitors Using Multitask Learning and Multiview Frameworks. ACS OMEGA 2024; 9:45159-45168. [PMID: 39554430 PMCID: PMC11561616 DOI: 10.1021/acsomega.4c06224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024]
Abstract
PI3K (phosphatidylinositol 3-kinase) is an intracellular phosphatidylinositol kinase composed of a regulatory subunit, p85, and a catalytic subunit, p110. Based on the different structures of the p110 catalytic subunit, PI3K can be divided into four isoforms: PI3Kα, PI3Kβ, PI3Kγ, and PI3Kδ. As molecularly targeted drugs, PI3K inhibitors have demonstrated antiproliferative effects on tumor cells and can also induce cancer cell death. In this study, a multiview deep learning framework (MVGNet) is proposed, which integrates fragment-based pharmacophore information and utilizes multitask learning to capture correlation information between subtasks. This framework predicts the inhibitory activity of molecules against the four PI3K isoforms (PI3Kα, PI3Kβ, PI3Kγ, and PI3Kδ). Compared to baseline prediction models based on three traditional machine learning methods (RF, SVM, and XGBoost) and four deep learning algorithms (GAT, D-MPNN, CMPNN, and KANO), our model demonstrates superior performance. The evaluation results show that our model achieves the highest average AUC-ROC and AUC-PR values on the test set, which are 0.927 ± 0.006 and 0.980 ± 0.002, respectively. This study provides a reference for exploring the structure-activity relationship of PI3K inhibitors.
Collapse
Affiliation(s)
- Yanlei Kang
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Re-sources, School of Information Engineering, Huzhou University, Huzhou 313000, Zhejiang Province,China
| | - Qiwei Xia
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Re-sources, School of Information Engineering, Huzhou University, Huzhou 313000, Zhejiang Province,China
| | - Yunliang Jiang
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Re-sources, School of Information Engineering, Huzhou University, Huzhou 313000, Zhejiang Province,China
- School of Computer Science and Technology, Zhejiang Normal University, Jinhua 321004, Zhejiang Province, China
| | - Zhong Li
- Zhejiang Province Key Laboratory of Smart Management & Application of Modern Agricultural Re-sources, School of Information Engineering, Huzhou University, Huzhou 313000, Zhejiang Province,China
| |
Collapse
|
33
|
Eun JR. Overview of hepatocarcinogenesis focusing on cellular origins of liver cancer stem cells: a narrative review. JOURNAL OF YEUNGNAM MEDICAL SCIENCE 2024; 42:3. [PMID: 39523770 PMCID: PMC11812091 DOI: 10.12701/jyms.2024.01088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Hepatocellular carcinoma (HCC) accounts for 85% to 90% of primary liver cancers and generally has a poor prognosis. The hierarchical model, which posits that HCC originates from liver cancer stem cells (CSCs), is now widely accepted, as it is for other cancer types. As CSCs typically reside in the G0 phase of the cell cycle, they are resistant to conventional chemotherapy. Therefore, to effectively treat HCC, developing therapeutic strategies that target liver CSCs is essential. Clinically, HCCs exhibit a broad spectrum of pathological and clinical characteristics, ranging from well-differentiated to poorly differentiated forms, and from slow-growing tumors to aggressive ones with significant metastatic potential. Some patients with HCC also show features of cholangiocarcinoma. This HCC heterogeneity may arise from the diverse cellular origins of liver CSCs. This review explores the normal physiology of liver regeneration and provides a comprehensive overview of hepatocarcinogenesis, including cancer initiation, isolation of liver CSCs, molecular signaling pathways, and microRNAs. Additionally, the cellular origins of liver CSCs are reviewed, emphasizing hematopoietic and mesenchymal stem cells, along with the well-known hepatocytes and hepatic progenitor cells.
Collapse
Affiliation(s)
- Jong Ryeol Eun
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Ilsan, Korea
| |
Collapse
|
34
|
Jiang Z, Xu Y, Yang L, Huang X, Bao J. Bile acid conjugated chitosan nanoparticles promote the proliferation and epithelial-mesenchymal transition of hepatocellular carcinoma by regulating the PI3K/Akt/mTOR pathway. Carbohydr Res 2024; 545:109296. [PMID: 39471534 DOI: 10.1016/j.carres.2024.109296] [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: 06/26/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
Bile acids have been known to play significant roles at certain physiological levels in gastrointestinal metabolism. Yet, they are known to be carcinogenic and aid in tumor progression in most cases, although the roles remain uncertain. Hence, we tested the cytotoxic potential of cholic acid (CA) loaded chitosan nanoparticles (CNPs) on Hep3B cells. The physicochemical properties of the CNPs synthesized with CA load (CA-CNPs) were determined using standard techniques such as ultraviolet-visible spectrophotometry (UV-Vis), fourier-transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), dynamic light scattering (DLS) and transmission electron microscopy (TEM). The characteristic peak for chitosan nanoparticles were observed for plain CNPs (pCNPs) and CA-CNPs at around 300 nm as per UV-Vis analysis. FTIR analysis indicated the possible trapping of CA onto CNPs as certain peaks were retained and some peaks were shifted. XRD analysis determined that the peaks representing CA and pCNPs were collectively obtained in CA-CNPs. As per DLS analysis, the particle size, PDI and ζ-potential of the CA-CNPs were 259 nm, 0.284 and 30.4 mV. Further, the CA-CNPs were non-cytotoxic on Hep3B cells at the maximum tested concentration of 500 μg/mL. The viability at 500 μg/mL of CA-CNPs was two-fold higher than 500 μg/mL of pCNPs. Also, the pCNPs were not hemolytic and therefore could not have played a role in the increase of viability after treatment with CA-CNPs, which indicates that CA posed a major role in increased viability of Hep3B cells. As per quantitative PCR (qPCR), the upregulated gene expressions of PI3K, Akt, mTORC2, cMyc, Fibronectin, hVPS34, Slug and ZEB1 and the downregulated expression of the tumor suppressor PTEN indicates that PI3K/Akt/mTOR pathway mediated the induction of epithelial-to-mesenchymal transition (EMT) in response to CA-CNPs treatment on Hep3B cells.
Collapse
Affiliation(s)
- Ziyu Jiang
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China; Department of Oncology, Lianyungang Hospital Affiliated to Xuzhou Medical University, Lianyungang, 222002, China
| | - Yi Xu
- Phase I Clinical Trial Center, Lianyungang Hospital Affiliated to Xuzhou Medical University, Lianyungang, 222002, China
| | - Liu Yang
- Department of Colorectal Center, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Xing Huang
- Department of Pathology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Affifiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Jun Bao
- Department of Chemotherapy, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China.
| |
Collapse
|
35
|
Jiang W, Wang Y, He W, Wang P, Meng P, Zhang S. CircCOCH plays a critical role in Hepatocellular carcinoma through modulating miR-450a and activating PI3K/mTOR pathway. Transl Oncol 2024; 49:102090. [PMID: 39181116 PMCID: PMC11388191 DOI: 10.1016/j.tranon.2024.102090] [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: 10/12/2023] [Revised: 07/29/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer with high pathogenicity and extremely poor prognosis. The role of circular RNAs (circRNAs) in HCC carcinogenesis and progression remains to be determined. Based on the analysis of HCC-related databases, as well as the expression analysis and identification of 25 HCC patient tissues and HCC cell lines, we found that the hsa_circ_0031431 (circCOCH) is significantly highly expressed in HCC tissues and cell lines. High circCOCH expression is associated with enhanced tumor proliferation and metastasis, and knocking down circCOCH can inhibit the growth of HCC in vivo and in vitro. Mechanistic studies show that circCOCH upregulates the expression of epidermal growth factor receptor (EGFR) through sponge miR-450a, thereby activating the Phosphoinositide 3-kinases (PI3Ks) cell pathway to promote HCC proliferation and metastasis. Futhermore, we found that IGF2BP3 mediates the biogenesis of circCOCH. The present study provides innovative insights into the role of circRNAs in the etiology of HCC carcinogenesis and might serve as a new promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Weiwei Jiang
- School of Medicine, Shangqiu Institute of Technology, Shangqiu, China
| | - Yan Wang
- Shangqiu Medical College, Shangqiu, China
| | - Wanli He
- School of Medicine, Shangqiu Institute of Technology, Shangqiu, China
| | - Peng Wang
- School of nursing and health, Zhengzhou University, Zhengzhou, China
| | - Peng Meng
- Department of Obstetrics and Gynecology, Xijing Hospital Affiliated to the Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Shanfeng Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
36
|
Gong X, Zhou Y, Wu P, He L, Ou C, Xiao X, Hou X, Shen Y, Li M, Tan Z, Xia X, Wang S. The petroleum ether extracts of Chloranthus fortunei(A. Gray) Solms-Laub.with bioactivities: A rising source in HCC drug treatment. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118414. [PMID: 38830451 DOI: 10.1016/j.jep.2024.118414] [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: 04/02/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hepatocellular Carcinoma (HCC) is an aggressive killer worldwide with high incidence and mortality. The herb Chloranthus fortunei (A. Gray) Solms-Laub is known as "Si Ji Feng" and is classified as a Feng-type medicine in classic Yao medicines. According to Yao's medical beliefs, Chloranthus fortunei has the functions of dispelling Feng, regulating qi, detoxifying, promoting blood circulation, etc. Folk uses its decoctions to treat stagnant liver conditions, such as liver abscesses, cirrhosis, hepatitis, and liver cancer. However, the bioactivity and mechanisms of Chloranthus fortunei extract against HCC have not been reported. AIM OF THE STUDY To investigate the anti-HCC bioactivity and potential mechanism of the extract of Chloranthus fortunei (CFS). MATERIALS AND METHODS Using 70% ethanol for reflux extraction of CFS resulted in the CFS ethanol extract, followed by sequential extractions with petroleum ether, chloroform, ethyl acetate, and n-butanol, yielding four fractions. The CCK-8 assay was utilized to examine the cytotoxic effects of 4 fractions on MHCC97-H and HepG2 cells, exploring the most effective component, namely petroleum ether extracts of CFS (PECFS). The major active ingredients of PECFS were identified using LC/MS technology, and the impact on cell proliferation and apoptosis in HCC cells was studied. The key genes and proteins in the pathway were validated using RT-PCR and Western blotting. BALB/c nude mice were chosen for tumor xenotransplantation and PECFS therapy. hinders the proliferation of HCC cells and promotes apoptosis. RESULTS Among the four fractions, it was found that PECFS have the highest antiproliferative activity against MHCC97-H and HepG2 cells (IC50 = 13.86, 10.55 μg/mL), with sesquiterpene compounds being the primary active constituents. The antiproliferative activity of PECFS on HCC cells was linked to the inhibition of cell cloning, invasion, and metastasis abilities, as well as the arrest of the cell cycle at the G2/M phase. Additionally, exerts pro-apoptotic effects on HCC cells by upregulating the pro-apoptotic protein Bax, downregulating the anti-apoptotic protein Bcl-2, and activating the expression of the Caspase family. Moreover, protein and m-RNA expression data showed that PECFS inhibits HCC cell proliferation and promotes apoptosis by regulating the PI3K/AKT/mTOR pathway. Besides, after PECFS treatment, tumor growth in nude mice was suppressed. CONCLUSION PECFS can inhibit the viability of HCC cells by acting on the PI3K/AKT/mTOR pathway, demonstrating anti-tumor potential. This study's findings suggest that PECFS may represent a promising source of novel agents for liver cancer treatment, providing scientific evidence for the traditional application of CFS in treating HCC.
Collapse
Affiliation(s)
- Xiaomei Gong
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China; National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Yun Zhou
- The School of Optometry & Ophthalmology, Tianjin Medical University, Tianjin, 300070, PR. China.
| | - Peiying Wu
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Lili He
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Chunli Ou
- Guangxi Key Laboratory of Medicinal Resources Protection and Genetic Improvement, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Xingyu Xiao
- Pharmaceutical College Guangxi Medical University, Nanning, 530021, PR.China.
| | - Xiaoli Hou
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Yuanyuan Shen
- China-ASEAN Traditional Medicine Exchange and Cooperation Centre, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Meng Li
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Zhien Tan
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Xianghua Xia
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| | - Shuo Wang
- National Engineering Research Center of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning, 530023, PR. China.
| |
Collapse
|
37
|
Ghnim ZS, Mahdi MS, Ballal S, Chahar M, Verma R, Al-Nuaimi AMA, Kumar MR, Al-Hussein RKA, Adil M, Jawad MJ. The role of kinesin superfamily proteins in hepatocellular carcinoma. Med Oncol 2024; 41:271. [PMID: 39400594 DOI: 10.1007/s12032-024-02497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024]
Abstract
The most prevalent form of primary liver cancer, hepatocellular carcinoma (HCC) poses a significant global health challenge due to its limited therapeutic options. Researchers are currently focused on the complex molecular landscape that governs the initiation and progression of HCC in order to identify new avenues for diagnosis, prognosis, and treatment. In the context of HCC, the Kinesin Superfamily Proteins (KIFs) have become critical regulators of cellular processes, prompting a growing interest in their function among the diverse array of molecular actors implicated in cancer. The KIFs, a family of microtubule-based molecular motors, are renowned for their essential roles in the dynamics of mitotic spindles and intracellular transport. Beyond their well-established functions in normal cellular physiology, emerging evidence indicates that dysregulation of KIFs significantly contributes to the pathogenesis of HCC. Novel therapeutic targets and diagnostic markers are revealed through the unique opportunity to comprehend the complex interplay between KIFs and the molecular events that drive HCC.
Collapse
Affiliation(s)
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University, Jaipur, Rajasthan, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Amritsar, Punjab, 140307, India
| | | | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | | | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
38
|
Sharma N, Singh L, Sharma A, Kumar A, Mahajan D. NAFLD-associated hepatocellular carcinoma (HCC) - A compelling case for repositioning of existing mTORc1 inhibitors. Pharmacol Res 2024; 208:107375. [PMID: 39209081 DOI: 10.1016/j.phrs.2024.107375] [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] [Received: 05/10/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The increasing prevalence of non-alcoholic fatty liver disease (NAFLD) is a growing concern for the high incidence rate of hepatocellular carcinoma (HCC) globally. The progression of NAFLD to HCC is heterogeneous and non-linear, involving intermediate stages of non-alcoholic steatohepatitis (NASH), liver fibrosis, and cirrhosis. There is a high unmet clinical need for appropriate diagnostic, prognostic, and therapeutic options to tackle this emerging epidemic. Unfortunately, at present, there is no validated marker to identify the risk of developing HCC in patients suffering from NAFLD or NASH. Additionally, the current treatment protocols for HCC don't differentiate between viral infection or NAFLD-specific etiology of the HCC and have a limited success rate. The mammalian target of rapamycin complex 1 (mTORc1) is an important protein involved in many vital cellular processes like lipid metabolism, glucose homeostasis, and inflammation. These cellular processes are highly implicated in NAFLD and its progression to severe liver manifestations. Additionally, hyperactivation of mTORc1 is known to promote cell proliferation, which can contribute to the genesis and progression of tumors. Many mTORc1 inhibitors are being evaluated for different types of cancers under various phases of clinical trials. This paper deliberates on the strong pathological implication of the mTORc1 signaling pathway in NAFLD and its progression to NASH and HCC and advocates for a systematic investigation of known mTORc1 inhibitors in suitable pre-clinical models of HCC having NAFLD/NASH-specific etiology.
Collapse
Affiliation(s)
- Nutan Sharma
- Center for Drug Discovery, BRIC-Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India; Department of Chemistry, Faculty of Applied and Basic Sciences, SGT University, Gurugram 122505, India
| | - Lakhwinder Singh
- Center for Drug Discovery, BRIC-Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India
| | - Aditya Sharma
- Center for Drug Discovery, BRIC-Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India
| | - Ajay Kumar
- Center for Drug Discovery, BRIC-Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India
| | - Dinesh Mahajan
- Center for Drug Discovery, BRIC-Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, India.
| |
Collapse
|
39
|
Liu R, Xue J, Han J, Tu M, Wang W, Chen Z, Qian X, Xiao B, Liang L. Cytarabine chemotherapy induces meibomian gland dysfunction. Ocul Surf 2024; 34:444-458. [PMID: 39395739 DOI: 10.1016/j.jtos.2024.10.002] [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: 07/01/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
PURPOSE Cytarabine (Ara-C) chemotherapy causes symptoms resembling meibomian gland dysfunction (MGD), suggesting potential associations between Ara-C and MGD. In this study, the pathological effects of Ara-C on MGD were investigated in a rodent model. METHODS Mice received Ara-C with or without rosiglitazone (PPARγ agonist) for 7 consecutive days. Slit-lamp biomicroscope was used for ocular examinations. Immunofluorescence detected acinar cell proliferation, differentiation, and ductal keratinization in the meibomian gland (MG). Lipid accumulation was evaluated by Oil Red O and LipidTox staining. Lipogenic status, FoxO1/FoxO3a cellular localization, and oxidative stress were visualized via immunohistochemistry. Western blotting assessed relative protein expression and AKT/FoxO1/FoxO3a pathway phosphorylation. RESULTS Ara-C (50 mg/kg) did not affect mouse survival but induced damage to ocular surface microenvironment, including corneal epithelial defects, MG orifice plugging and acinar dropout, and lacrimal gland (LG) dysfunction. Ara-C intervention inhibited proliferation and caused progenitor loss in the MG, as evidenced by reduced PCNA + labeling and P63+/Lrig1+ basal cell numbers. The MG ducts of Ara-C-treated mice exhibited marked dilatation, lipid deposition, and hyperkeratinization (K1/K10 overexpression). Ara-C disrupted MG lipid metabolism by downregulating PPARγ and its downstream lipogenic targets AWAT2/SOAT1/ELOVL4 and upregulating HMGCR. Dephosphorylation of AKT and the subsequent nuclear translocation of FoxO1/FoxO3a contributed to Ara-C-induced PPARγ downregulation. Ara-C triggered oxidative stress with increases in 4-HNE and 8-OHdG and Keap1/Nrf2/HO-1/SOD1 axis dysregulation. Rosiglitazone treatment ameliorated MGD-associated pathological manifestations, LG function, MG lipid metabolism, and oxidative stress in Ara-C-exposed mice. CONCLUSIONS Systemic Ara-C chemotherapy exerted topical cytotoxic effects on the ocular surface, and PPARγ restoration by rosiglitazone mitigated Ara-C-induced MGD alterations.
Collapse
Affiliation(s)
- Ren Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jianwen Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jiaxu Han
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mengqian Tu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Wenhui Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Ziyan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xiaobing Qian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Bing Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Lingyi Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
40
|
Zhu S, Jin Q, Zhang S, Song Z, Zhang S, Zhao Z. Integrating Network Pharmacology and Experimental Verification to Explore the Pharmacological Mechanisms of Radix Paeoniae Rubra Against Glioma. Appl Biochem Biotechnol 2024; 196:6424-6441. [PMID: 38381309 DOI: 10.1007/s12010-024-04887-6] [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: 02/12/2024] [Indexed: 02/22/2024]
Abstract
Glioma has a high mortality and can hardly be completely cured. Radix Paeoniae Rubra (RPR) is a prevalent component in traditional Chinese medicine used for tumor treatments. We explored the mechanism of RPR in treating glioma using network pharmacology and experiments. A network pharmacology approach was used to screen active ingredients, targets of RPR and glioma. We then constructed a herb-active ingredient-target-pathway network and conducted protein-protein interaction (PPI) network analysis, as well as Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Molecular docking was also performed. Using CCK-8, colony formation, and xenograft experiments, we evaluated the effect of RPR on glioma. The involved pathway and proteins were identified by Western blot. From public databases, we identified nine active RPR ingredients and 40 overlapping targets among 109 RPR targets and 1360 glioma-associated targets. The PPI analysis revealed ten targets, such as AKT1, TP53, and VEGFA, which were identified as hub genes. The results from GO and KEGG analysis highlighted the involvement of the PI3K/AKT pathway. A herb-active ingredient-target-pathway network was constructed. By docking molecular structures, six suitable conformations have been identified. The RPR extract demonstrated anti-tumor properties by inhibiting glioma cell proliferation in vitro and in vivo, likely achieved by suppressing the phosphorylation of the PI3K/AKT signaling pathway. RPR concurrently downregulated the phosphorylation level of AKT1 and the protein expression level of VEGFA, while upregulating the expression of P53 in the U251 cell line. Utilizing network pharmacology and molecular docking, our study not only predicted the impact of RPR on glioma but also delineated the herb-active ingredient-target-pathway network. Experimentally, we confirmed that RPR may exert its anti-tumor properties by inhibiting the phosphorylation of the PI3K/AKT pathway, including AKT1, and by regulating the expression levels of VEGFA and P53.
Collapse
Affiliation(s)
- Siyu Zhu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qianxu Jin
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiyang Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zihan Song
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiqi Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
41
|
Kreis NN, Moon HH, Wordeman L, Louwen F, Solbach C, Yuan J, Ritter A. KIF2C/MCAK a prognostic biomarker and its oncogenic potential in malignant progression, and prognosis of cancer patients: a systematic review and meta-analysis as biomarker. Crit Rev Clin Lab Sci 2024; 61:404-434. [PMID: 38344808 PMCID: PMC11815995 DOI: 10.1080/10408363.2024.2309933] [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: 10/31/2023] [Revised: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 03/24/2024]
Abstract
KIF2C/MCAK (KIF2C) is the most well-characterized member of the kinesin-13 family, which is critical in the regulation of microtubule (MT) dynamics during mitosis, as well as interphase. This systematic review briefly describes the important structural elements of KIF2C, its regulation by multiple molecular mechanisms, and its broad cellular functions. Furthermore, it systematically summarizes its oncogenic potential in malignant progression and performs a meta-analysis of its prognostic value in cancer patients. KIF2C was shown to be involved in multiple crucial cellular processes including cell migration and invasion, DNA repair, senescence induction and immune modulation, which are all known to be critical during the development of malignant tumors. Indeed, an increasing number of publications indicate that KIF2C is aberrantly expressed in multiple cancer entities. Consequently, we have highlighted its involvement in at least five hallmarks of cancer, namely: genome instability, resisting cell death, activating invasion and metastasis, avoiding immune destruction and cellular senescence. This was followed by a systematic search of KIF2C/MCAK's expression in various malignant tumor entities and its correlation with clinicopathologic features. Available data were pooled into multiple weighted meta-analyses for the correlation between KIF2Chigh protein or gene expression and the overall survival in breast cancer, non-small cell lung cancer and hepatocellular carcinoma patients. Furthermore, high expression of KIF2C was correlated to disease-free survival of hepatocellular carcinoma. All meta-analyses showed poor prognosis for cancer patients with KIF2Chigh expression, associated with a decreased overall survival and reduced disease-free survival, indicating KIF2C's oncogenic potential in malignant progression and as a prognostic marker. This work delineated the promising research perspective of KIF2C with modern in vivo and in vitro technologies to further decipher the function of KIF2C in malignant tumor development and progression. This might help to establish KIF2C as a biomarker for the diagnosis or evaluation of at least three cancer entities.
Collapse
Affiliation(s)
- Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Ha Hyung Moon
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Frank Louwen
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
42
|
Liu J, Lu Y, Zheng B, Huang D, Song J, Wang B, Zheng S. Talin1 promotes HCC progression by regulating NRG1/PI3K/AKT pathway. Discov Oncol 2024; 15:360. [PMID: 39162903 PMCID: PMC11335986 DOI: 10.1007/s12672-024-01243-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 08/14/2024] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVE OF THE STUDY Hepatocellular carcinoma (HCC) stands as the third leading cause of cancer-related mortality globally. Metastasis, responsible for treatment failures, underscores the urgency to comprehend molecular drivers of invasion and migration. Central to the invasive and migratory processes underlying metastasis is the protein Talin1. However, the role and underlying mechanisms governing Talin1's involvement in HCC have remained elusive. METHODS A total of 100 HCC specimens were collected from patients who underwent hepatectomy in our center. The expression level of talin1 was measured to evaluate the correlationship of talin1 and the development of HCC. In vitro and in vivo experiments were conducted to verify the characteristic of talin1 in HCC. RNA-seq and bioinformatics analysis were performed to identify the downstream signal pathway of talin1 and their impact on HCC development. RESULTS Here, we reported elevated levels of Talin1 mRNA and protein in HCC tissues. Meanwhile, downregulation of Talin1 significantly reduced the HCC cell proliferation and metastasis in vitro and in vivo. Furthermore, elevating NRG-1, a downstream target of Talin1, enhanced metastasis of HCC cells. More importantly, attenuation of Talin1 inhibited HCC progression through decreasing the stabilization of NRG1 mRNA, consequently regulating the expression of NRG1 and its involvement in mediating the PI3K/AKT pathway. CONCLUSION Taken together, Talin1 regulates cellular proliferation, metastasis, and invasiveness by modulating NRG1/PI3K/AKT axis, suggesting that Talin1 emerges as a promising candidate for treating HCC.
Collapse
Affiliation(s)
- Jialong Liu
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yao Lu
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bowen Zheng
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Deng Huang
- Department of Hepatobiliary, General Hospital of Tibet Military Command Area, Lhasa, Tibet, China
| | - Juxian Song
- Department of Hepatobiliary Surgery, The 925th Hospital of the PLA, Guiyang, Guizhou, China
| | - Baolin Wang
- Department of Surgery, The 63650th Troop Hospital of the Chinese People's Liberation Army, Korla, Xinjiang, China
| | - Shuguo Zheng
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
43
|
Elbaset MA, Afifi SM, Esatbeyoglu T, Abdelrahman SS, Saleh DO. Neuroprotective Effects of Trimetazidine against Cisplatin-Induced Peripheral Neuropathy: Involvement of AMPK-Mediated PI3K/mTOR, Nrf2, and NF- κB Signaling Axes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:6612009. [PMID: 39502494 PMCID: PMC11535264 DOI: 10.1155/2024/6612009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 05/28/2024] [Accepted: 07/06/2024] [Indexed: 11/08/2024]
Abstract
Cisplatin-induced peripheral neuropathy (CIPN) is a common and debilitating side effect of cisplatin chemotherapy used in cancer treatment. This study explored the neuroprotective effects of Trimetazidine (TRI) against CIPN by preserving nerve integrity, reducing neuro-oxidative stress, and alleviating neuroinflammation. Using a rat model of CIPN, we evaluated TRI's impact on motor coordination, pain sensitivity, and peripheral nerve histopathology. Also, its effects on neuro-oxidative stress and neuroinflammatory markers were assessed. The findings showed that rats with CIPN had worse motor coordination and increased sensitivity to pain but that these symptoms were alleviated by TRI therapy in a dose-dependent way. Nerve conduction velocities were normalized, and expression of genes involved in neuropathy signaling was suppressed after TRI therapy. Antioxidant benefits were also shown in TRI, with oxidative damage being reduced and the cellular energy balance being restored. By inhibiting the production of inflammatory markers, it also demonstrated anti-inflammatory properties. Histopathological examination revealed that TRI, especially when administered at a higher dose, inhibited the degeneration and demyelination of nerve fibers. The anti-inflammatory properties of TRI in the sciatic nerves were further shown by the fact that its administration reduced iNOS expression. In conclusion, AMPK-mediated PI3K/mTOR, Nrf2, and NF-κB signaling pathways may all be involved in the therapeutic benefits of TRI for CIPN. These results indicate that TRI may be useful for reducing the side effects of CIPN and enhancing patient outcomes during cisplatin chemotherapy.
Collapse
Affiliation(s)
- Marawan A. Elbaset
- Pharmacology DepartmentMedical Research and Clinical Studies InstituteNational Research Centre, 33 El-Bohouth Street, Dokki, Cairo P.O. 12622, Egypt
| | - Sherif M. Afifi
- Department for Life Quality StudiesRimini CampusUniversity of Bologna, Corso d'Augusto, 237, Rimini 47921, Italy
| | - Tuba Esatbeyoglu
- Department of Molecular Food Chemistry and Food DevelopmentInstitute of Food and One HealthGottfried Wilhelm Leibniz University, Am Kleinen Felde 30, Hannover 30167, Germany
| | - Sahar S. Abdelrahman
- Department of PathologyCollege of Veterinary MedicineCairo University, Cairo P.O. 12211, Egypt
| | - Dalia O. Saleh
- Pharmacology DepartmentMedical Research and Clinical Studies InstituteNational Research Centre, 33 El-Bohouth Street, Dokki, Cairo P.O. 12622, Egypt
| |
Collapse
|
44
|
Valdez-Salazar F, Jiménez-Del Rio LA, Padilla-Gutiérrez JR, Valle Y, Muñoz-Valle JF, Valdés-Alvarado E. Advances in Melanoma: From Genetic Insights to Therapeutic Innovations. Biomedicines 2024; 12:1851. [PMID: 39200315 PMCID: PMC11351162 DOI: 10.3390/biomedicines12081851] [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: 06/14/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Advances in melanoma research have unveiled critical insights into its genetic and molecular landscape, leading to significant therapeutic innovations. This review explores the intricate interplay between genetic alterations, such as mutations in BRAF, NRAS, and KIT, and melanoma pathogenesis. The MAPK and PI3K/Akt/mTOR signaling pathways are highlighted for their roles in tumor growth and resistance mechanisms. Additionally, this review delves into the impact of epigenetic modifications, including DNA methylation and histone changes, on melanoma progression. The tumor microenvironment, characterized by immune cells, stromal cells, and soluble factors, plays a pivotal role in modulating tumor behavior and treatment responses. Emerging technologies like single-cell sequencing, CRISPR-Cas9, and AI-driven diagnostics are transforming melanoma research, offering precise and personalized approaches to treatment. Immunotherapy, particularly immune checkpoint inhibitors and personalized mRNA vaccines, has revolutionized melanoma therapy by enhancing the body's immune response. Despite these advances, resistance mechanisms remain a challenge, underscoring the need for combined therapies and ongoing research to achieve durable therapeutic responses. This comprehensive overview aims to highlight the current state of melanoma research and the transformative impacts of these advancements on clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Emmanuel Valdés-Alvarado
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.V.-S.)
| |
Collapse
|
45
|
Kassem PH, Montasser IF, Mahmoud RM, Ghorab RA, AbdelHakam DA, Fathi MESA, Wahed MAA, Mohey K, Ibrahim M, Hadidi ME, Masssoud YM, Salah M, Abugable A, Bahaa M, Khamisy SE, Meteini ME. Genomic landscape of hepatocellular carcinoma in Egyptian patients by whole exome sequencing. BMC Med Genomics 2024; 17:202. [PMID: 39123171 PMCID: PMC11311965 DOI: 10.1186/s12920-024-01965-w] [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: 02/02/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Chronic hepatitis and liver cirrhosis lead to accumulation of genetic alterations driving HCC pathogenesis. This study is designed to explore genomic landscape of HCC in Egyptian patients by whole exome sequencing. METHODS Whole exome sequencing using Ion Torrent was done on 13 HCC patients, who underwent surgical intervention (7 patients underwent living donor liver transplantation (LDLT) and 6 patients had surgical resection}. RESULTS Mutational signature was mostly S1, S5, S6, and S12 in HCC. Analysis of highly mutated genes in both HCC and Non-HCC revealed the presence of highly mutated genes in HCC (AHNAK2, MUC6, MUC16, TTN, ZNF17, FLG, MUC12, OBSCN, PDE4DIP, MUC5b, and HYDIN). Among the 26 significantly mutated HCC genes-identified across 10 genome sequencing studies-in addition to TCGA, APOB and RP1L1 showed the highest number of mutations in both HCC and Non-HCC tissues. Tier 1, Tier 2 variants in TCGA SMGs in HCC and Non-HCC (TP53, PIK3CA, CDKN2A, and BAP1). Cancer Genome Landscape analysis revealed Tier 1 and Tier 2 variants in HCC (MSH2) and in Non-HCC (KMT2D and ATM). For KEGG analysis, the significantly annotated clusters in HCC were Notch signaling, Wnt signaling, PI3K-AKT pathway, Hippo signaling, Apelin signaling, Hedgehog (Hh) signaling, and MAPK signaling, in addition to ECM-receptor interaction, focal adhesion, and calcium signaling. Tier 1 and Tier 2 variants KIT, KMT2D, NOTCH1, KMT2C, PIK3CA, KIT, SMARCA4, ATM, PTEN, MSH2, and PTCH1 were low frequency variants in both HCC and Non-HCC. CONCLUSION Our results are in accordance with previous studies in HCC regarding highly mutated genes, TCGA and specifically enriched pathways in HCC. Analysis for clinical interpretation of variants revealed the presence of Tier 1 and Tier 2 variants that represent potential clinically actionable targets. The use of sequencing techniques to detect structural variants and novel techniques as single cell sequencing together with multiomics transcriptomics, metagenomics will integrate the molecular pathogenesis of HCC in Egyptian patients.
Collapse
Affiliation(s)
- Perihan Hamdy Kassem
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Iman Fawzy Montasser
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Ramy Mohamed Mahmoud
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rasha Ahmed Ghorab
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dina A AbdelHakam
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Marwa A Abdel Wahed
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Khaled Mohey
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mariam Ibrahim
- Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed El Hadidi
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham Dubai Campus, Dubai, United Arab Emirates
- Bioinformatics Group, Center for Informatics Science(CIS), School of Information Technology and Computer Science(ITCS), Nile University, Giza, Egypt
| | - Yasmine M Masssoud
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Manar Salah
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Arwa Abugable
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Mohamad Bahaa
- Hepato-Pancreatico-Biliary Surgery Department and liver Transplantation, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Mahmoud El Meteini
- Hepato-Pancreatico-Biliary Surgery Department and liver Transplantation, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
46
|
Martucci NJ, Stoops J, Bowen W, Orr A, Cotner MC, Michalopoulos GK, Bhushan B, Mars WM. A Novel Role for the Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Delta Isoform in Hepatocellular Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1511-1527. [PMID: 38705383 PMCID: PMC11393825 DOI: 10.1016/j.ajpath.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024]
Abstract
The phosphatidylinositol-4,5-bisphosphate 3-kinase delta isoform (Pik3cd), usually considered immune-specific, was unexpectedly identified as a gene potentially related to either regeneration and/or differentiation in animals lacking hepatocellular Integrin Linked Kinase (ILK). Since a specific inhibitor (Idelalisib, or CAL101) for the catalytic subunit encoded by Pik3cd (p110δ) has reported hepatotoxicity when used for treating chronic lymphocytic leukemia and other lymphomas, the authors aimed to elucidate whether there is a role for p110δ in normal liver function. To determine the effect on normal liver regeneration, partial hepatectomy (PHx) was performed using mice in which p110δ was first inhibited using CAL101. Inhibition led to over a 50% decrease in proliferating hepatocytes in the first 2 days after PHx. This difference correlated with phosphorylation changes in the HGF and EGF receptors (MET and EGFR, respectively) and NF-κB signaling. Ingenuity Pathway Analyses implicated C/EBPβ, HGF, and the EGFR heterodimeric partner, ERBB2, as three of the top 20 regulators downstream of p110δ signaling because their pathways were suppressed in the presence of CAL101 at 1 day post-PHx. A regulatory role for p110δ signaling in mouse and rat hepatocytes through MET and EGFR was further verified using hepatocyte primary cultures, in the presence or absence of CAL101. Combined, these data support a role for p110δ as a downstream regulator of normal hepatocytes when stimulated to proliferate.
Collapse
Affiliation(s)
- Nicole J Martucci
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Stoops
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William Bowen
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne Orr
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary-Claire Cotner
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
47
|
Xiao F, Zhang Z, Li L, He X, Chen Y. LINC01370 suppresses hepatocellular carcinoma proliferation and metastasis by regulating the PI3K/AKT pathway. Discov Oncol 2024; 15:326. [PMID: 39090419 PMCID: PMC11294307 DOI: 10.1007/s12672-024-01193-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) poses a serious threat to human health worldwide. lncRNA dysregulation is frequently observed in various cancers, including HCC. However, the function of LINC01370 in HCC progression and its underlying mechanisms remain unclear. METHODS LINC01370 expression in HCC tissues with cells was analyzed by applying the GEO and GEPIA databases and qRT-PCR. CCK-8 and Transwell assays were used to assess HCC cell proliferation, migration, and invasion. The PI3K, AKT, with p-AKT protein expression were analyzed by western blotting. RESULTS Gene Expression Omnibus (GEO) and Gene Expression Profiling Interactive Analysis (GEPIA) showed that LINC01370 expression was significantly lower in HCC tissues than in normal tissues. LINC01370 overexpression markedly repressed HepG2 SMMC-7721 cells proliferation, migration, and invasion. To understand the downstream mechanism of LINC01370 regulation, we further analyzed the genes co-expressed with LINC01370 in GSE136247 and GSE132037 and then performed KEGG analysis. The PA pathway was found to be a downstream pathway regulated by LINC01370 in GSE136247 and GSE132037 via gene co-expression and KEGG analysis. Furthermore, PI3K and p-AKT protein levels decreased after LINC01370 overexpression. Importantly, rescue experiments showed that activation of the PI3K/AKT pathway disrupted the repressive effect of LINC01370 overexpression on the proliferation, migration, and invasion of HepG2 of SMMC-7721 cells. CONCLUSIONS This study verified that LINC01370 suppresses HCC proliferation with metastasis by regulating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Fei Xiao
- Laboratory Department, Maoming People's Hospital, No. 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Zhuoyun Zhang
- Cancer Department, Maoming People's Hospital, Maoming, 525000, China
| | - Luqian Li
- Laboratory Department, Maoming People's Hospital, No. 101 Weimin Road, Maoming, 525000, Guangdong, China
| | - Xiaojie He
- Department of Endocrinology, Maoming People's Hospital, Maoming, 525000, China
| | - Yufeng Chen
- Laboratory Department, Maoming People's Hospital, No. 101 Weimin Road, Maoming, 525000, Guangdong, China.
| |
Collapse
|
48
|
Dong L, Luo L, Wang Z, Lian S, Wang M, Wu X, Fan J, Zeng Y, Li S, Lv S, Yang Y, Chen R, Shen E, Yang W, Li C, Wang K. Targeted degradation of NDUFS1 by agrimol B promotes mitochondrial ROS accumulation and cytotoxic autophagy arrest in hepatocellular carcinoma. Free Radic Biol Med 2024; 220:111-124. [PMID: 38697493 DOI: 10.1016/j.freeradbiomed.2024.04.242] [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] [Received: 03/04/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Hepatocellular carcinoma (HCC) is a global public health problem with increased morbidity and mortality. Agrimol B, a natural polyphenol, has been proved to be a potential anticancer drug. Our recent report showed a favorable anticancer effect of agrimol B in HCC, however, the mechanism of action remains unclear. Here, we found agrimol B inhibits the growth and proliferation of HCC cells in vitro as well as in an HCC patient-derived xenograft (PDX) model. Notably, agrimol B drives autophagy initiation and blocks autophagosome-lysosome fusion, resulting in autophagosome accumulation and autophagy arrest in HCC cells. Mechanistically, agrimol B downregulates the protein level of NADH:ubiquinone oxidoreductase core subunit S1 (NDUFS1) through caspase 3-mediated degradation, leading to mitochondrial reactive oxygen species (mROS) accumulation and autophagy arrest. NDUFS1 overexpression partially restores mROS overproduction, autophagosome accumulation, and growth inhibition induced by agrimol B, suggesting a cytotoxic role of agrimol B-induced autophagy arrest in HCC cells. Notably, agrimol B significantly enhances the sensitivity of HCC cells to sorafenib in vitro and in vivo. In conclusion, our study uncovers the anticancer mechanism of agrimol B in HCC involving the regulation of oxidative stress and autophagy, and suggests agrimol B as a potential therapeutic drug for HCC treatment.
Collapse
Affiliation(s)
- Lixia Dong
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, PR China
| | - Zihao Wang
- Colorectal Cancer Center, West China Hospital, Sichuan University, 610041, PR China
| | - Shan Lian
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Mao Wang
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Xingyun Wu
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Jiawu Fan
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yan Zeng
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Sijia Li
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Sinan Lv
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yurong Yang
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Rong Chen
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Enhao Shen
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Wenyong Yang
- Department of Neurosurgery, Medical Research Center, the Third People's Hospital of Chengdu, the Affiliated Hospital of Southwest Jiaotong University, the Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, 610041, PR China.
| | - Changlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
49
|
Safri F, Nguyen R, Zerehpooshnesfchi S, George J, Qiao L. Heterogeneity of hepatocellular carcinoma: from mechanisms to clinical implications. Cancer Gene Ther 2024; 31:1105-1112. [PMID: 38499648 PMCID: PMC11327108 DOI: 10.1038/s41417-024-00764-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common types of primary liver cancer. Current treatment options have limited efficacy against this malignancy, primarily owing to difficulties in early detection and the inherent resistance to existing drugs. Tumor heterogeneity is a pivotal factor contributing significantly to treatment resistance and recurrent manifestations of HCC. Intratumoral heterogeneity is an important aspect of the spectrum of complex tumor heterogeneity and contributes to late diagnosis and treatment failure. Therefore, it is crucial to thoroughly understand the molecular mechanisms of how tumor heterogeneity develops. This review aims to summarize the possible molecular dimensions of tumor heterogeneity with an emphasis on intratumoral heterogeneity, evaluate its profound impact on the diagnosis and therapeutic strategies for HCC, and explore the suitability of appropriate pre-clinical models that can be used to best study tumor heterogeneity; thus, opening new avenues for cancer treatment.
Collapse
Affiliation(s)
- Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Shadi Zerehpooshnesfchi
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
50
|
Wei T, Cheng J, Ji Y, Cao X, Ding S, Liu Q, Wang Z. Baculovirus-mediated endostatin and angiostatin activation of autophagy through the AMPK/AKT/mTOR pathway inhibits angiogenesis in hepatocellular carcinoma. Open Life Sci 2024; 19:20220914. [PMID: 39091624 PMCID: PMC11291770 DOI: 10.1515/biol-2022-0914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 08/04/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly vascularized carcinoma, and targeting its neovascularization represents an effective therapeutic approach. Our previous study demonstrated that the baculovirus-mediated endostatin and angiostatin fusion protein (BDS-hEA) effectively inhibits the angiogenesis of vascular endothelial cells and the growth of HCC tumors. However, the mechanism underlying its anti-angiogenic effect remains unclear. Increasing evidence suggests that autophagy has a significant impact on the function of vascular endothelial cells and response to cancer therapy. Hence, the objective of this research was to investigate the correlation between BDS-hEA-induced angiogenesis inhibition and autophagy, along with potential regulatory mechanisms. Our results demonstrated that BDS-hEA induced autophagy in EA.hy926 cells, as evidenced by the increasing number of autophagosomes and reactive oxygen species, accompanied by an upregulation of Beclin-1, LC3-II/LC3-I, and p62 protein expression. Suppression of autophagy using 3-methyladenine attenuated the functions of BDS-hEA-induced EA.hy926 cells, including the viability, proliferation, invasion, migration, and angiogenesis. Moreover, BDS-hEA induced autophagy by downregulating the expression of CD31, VEGF, and VEGFR2, as well as phosphorylated protein kinase B (p-AKT) and phosphorylated mammalian target of rapamycin (p-mTOR), while concurrently upregulating phosphorylated AMP-activated protein kinase (p-AMPK). The in vivo results further indicated that inhibition of autophagy by chloroquine significantly impeded the ability of BDS-hEA to suppress HCC tumor growth in mice. Mechanistically, BDS-hEA prominently facilitated autophagic apoptosis in tumor tissues and decreased the levels of ki67, CD31, VEGF, MMP-9, p-AKT, and p-mTOR while simultaneously enhancing the p-AMPK expression. In conclusion, our findings suggest that BDS-hEA induces autophagy as a cytotoxic response by modulating the AMPK/AKT/mTOR signaling pathway, thereby exerting anti-angiogenic effects against HCC.
Collapse
Affiliation(s)
- Tingting Wei
- Department of Oncology, General Hospital of Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750001, P.R. China
| | - Jiajie Cheng
- School of Pharmacy, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, P. R. China
| | - Yonggan Ji
- School of Pharmacy, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, P. R. China
| | - Xue Cao
- Department of Oncology, General Hospital of Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750001, P.R. China
| | - Shuqin Ding
- School of Inspection, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, P.R. China
| | - Quanxia Liu
- Department of Oncology, General Hospital of Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750001, P.R. China
| | - Zhisheng Wang
- School of Inspection, Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, P.R. China
| |
Collapse
|