1
|
Zhou C, Zhong R, Zhang L, Yang R, Luo Y, Lei H, Li L, Cao J, Yuan Z, Tan X, Xie M, Qu H, He Z. Exploring the mechanism of rosmarinic acid in the treatment of lung adenocarcinoma based on bioinformatics methods and experimental validation. Discov Oncol 2025; 16:47. [PMID: 39812944 PMCID: PMC11735722 DOI: 10.1007/s12672-025-01784-0] [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: 08/10/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Rosmarinic acid (RosA) is a natural polyphenol compound that has been shown to be effective in the treatment of inflammatory disease and a variety of malignant tumors. However, its specific mechanism for the treatment of lung adenocarcinoma (LUAD) has not been fully elucidated. Therefore, this study aims to clarify the mechanism of RosA in the treatment of LUAD by integrating bioinformatics, network pharmacology and in vivo experiments, and to explore the potential of the active ingredients of traditional Chinese medicine in treating LUAD. METHODS Firstly, the network pharmacology was used to screen the RosA targets, and LUAD-related differential expressed genes (DEGs) were acquired from the GEO database. The intersection of LUAD regulated by RosA (RDEGs) was obtained through the Venn diagram. Secondly, GO and KEGG enrichment analysis of RDEGs were performed, and protein-protein interaction networks (PPIs) were constructed to identify and visualize hub RDEGs. Then, molecular docking between hub RDEGs and RosA was performed, and further evaluation was carried out by using bioinformatics for the predictive value of the hub RDEGs. Finally, the mechanism of RosA in the treatment of LUAD was verified by establishing a xenograft model of NSCLC in nude mouse. RESULTS Bioinformatics and other analysis showed that, compared with the control group, the expressions of MMP-1, MMP-9, IGFBP3 and PLAU in LUAD tissues were significantly up-regulated, and the expressions of PPARG and FABP4 were significantly down-regulated, and these hub RDEGs had potential predictive value for LUAD. In vivo experimental results showed that RosA could inhibit the growth of transplanted tumors in nude mice bearing tumors of lung cancer cells, reduce the positive expression of Ki67 in lung tumor tissue, and hinder the proliferation of lung tumor cells. Upregulated expression of PPARG and FABP4 by activating the PPAR signaling pathway increases the level of ROS in lung tumor tissues and promotes apoptosis of lung tumor cells. In addition, RosA can also reduce the expression of MMP-9 and IGFBP3, inhibit the migration and invasion of lung tumor tissue cells. CONCLUSIONS This study demonstrated that RosA could induce apoptosis by regulating the PPAR signaling pathway and the expression of MMP-9, inhibit the proliferation, migration and invasion of lung cancer cells, thereby exerting anti-LUAD effects. This study provides new insight into the potential mechanism of RosA in treating LUAD and provides a new therapeutic avenue for treatment of LUAD.
Collapse
Affiliation(s)
- Chaowang Zhou
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Ruqian Zhong
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Lei Zhang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Renyi Yang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Yuxin Luo
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Huijun Lei
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Liang Li
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Jianzhong Cao
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Zhiying Yuan
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Xiaoning Tan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China
| | - Mengzhou Xie
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Haoyu Qu
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Zuomei He
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China.
| |
Collapse
|
2
|
Li H, Zhang L, Shu B, Wang X, Yang S. Endoplasmic reticulum stress-related signatures: a game-changer in prognostic stratification for hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2024:00042737-990000000-00443. [PMID: 39589828 DOI: 10.1097/meg.0000000000002894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has limited therapeutic options and a poor prognosis. The endoplasmic reticulum (ER) plays a crucial role in tumor progression and response to stress, making it a promising target for HCC stratification. This study aimed to develop a risk stratification model using ER stress-related signatures. METHODS We utilized transcriptome data from The Cancer Genome Atlas and Gene Expression Omnibus, which encompass whole-genome expression profiles and clinical annotations. Machine learning algorithms, including the least absolute shrinkage and selection operator, random forest, and support vector machine recursive feature elimination, were applied to the key genes associated with HCC prognosis. A prognostic system was developed using univariate Cox hazard analysis and least absolute shrinkage and selection operator Cox regression, followed by validation using Kaplan-Meier analysis and receiver operating characteristic curves. Tumor immune dysfunction and exclusion tools were used to predict immunotherapy responsiveness. RESULTS Two distinct clusters associated with ER stress were identified in HCC, each exhibiting unique clinical and biological features. Using a computational approach, a prognostic risk model, namely the ER stress-related signature, was formulated, demonstrating enhanced predictive accuracy compared with that of existing prognostic models. An effective clinical nomogram was established by integrating the risk model with clinicopathological factors. Patients with lower risk scores exhibited improved responsiveness to various chemotherapeutic, targeted, and immunotherapeutic agents. CONCLUSION The critical role of ER stress in HCC is highlighted. The ER stress-related signature developed in this study is a powerful tool to assess the risk and clinical treatment of HCC.
Collapse
Affiliation(s)
| | - Lei Zhang
- Department of Ultrasound, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University
| | - Bin Shu
- Hepatopancereatobiliary Center
| | | | - Shizhong Yang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Institute for Precision Medicine, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), Tsinghua University
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Chen M, Wang H, Cui Q, Shi J, Hou Y. Dual function of activated PPARγ by ligands on tumor growth and immunotherapy. Med Oncol 2024; 41:114. [PMID: 38619661 DOI: 10.1007/s12032-024-02363-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
As one of the peroxisome-proliferator-activated receptors (PPARs) members, PPARγ is a ligand binding and activated nuclear hormone receptor, which is an important regulator in metabolism, proliferation, tumor progression, and immune response. Increased evidence suggests that activation of PPARγ in response to ligands inhibits multiple types of cancer proliferation, metastasis, and tumor growth and induces cell apoptosis including breast cancer, colon cancer, lung cancer, and bladder cancer. Conversely, some reports suggest that activation of PPARγ is associated with tumor growth. In addition to regulating tumor progression, PPARγ could promote or inhibit tumor immunotherapy by affecting macrophage differentiation or T cell activity. These controversial findings may be derived from cancer cell types, conditions, and ligands, since some ligands are independent of PPARγ activity. Therefore, this review discussed the dual role of PPARγ on tumor progression and immunotherapy.
Collapse
Affiliation(s)
- Mingjun Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Huijie Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Qian Cui
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China.
| |
Collapse
|
4
|
Zhang J, Tang M, Shang J. PPARγ Modulators in Lung Cancer: Molecular Mechanisms, Clinical Prospects, and Challenges. Biomolecules 2024; 14:190. [PMID: 38397426 PMCID: PMC10886696 DOI: 10.3390/biom14020190] [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: 12/30/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Lung cancer is one of the most lethal malignancies worldwide. Peroxisome proliferator-activated receptor gamma (PPARγ, NR1C3) is a ligand-activated transcriptional factor that governs the expression of genes involved in glucolipid metabolism, energy homeostasis, cell differentiation, and inflammation. Multiple studies have demonstrated that PPARγ activation exerts anti-tumor effects in lung cancer through regulation of lipid metabolism, induction of apoptosis, and cell cycle arrest, as well as inhibition of invasion and migration. Interestingly, PPARγ activation may have pro-tumor effects on cells of the tumor microenvironment, especially myeloid cells. Recent clinical data has substantiated the potential of PPARγ agonists as therapeutic agents for lung cancer. Additionally, PPARγ agonists also show synergistic effects with traditional chemotherapy and radiotherapy. However, the clinical application of PPARγ agonists remains limited due to the presence of adverse side effects. Thus, further research and clinical trials are necessary to comprehensively explore the actions of PPARγ in both tumor and stromal cells and to evaluate the in vivo toxicity. This review aims to consolidate the molecular mechanism of PPARγ modulators and to discuss their clinical prospects and challenges in tackling lung cancer.
Collapse
Affiliation(s)
- Jiyun Zhang
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 511436, China;
- Guangzhou National Laboratory, Guangzhou 510005, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Miru Tang
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Jinsai Shang
- School of Basic Medical Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou 511436, China;
- Guangzhou National Laboratory, Guangzhou 510005, China
| |
Collapse
|
5
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [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: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Hounye AH, Hu B, Wang Z, Wang J, Cao C, Zhang J, Hou M, Qi M. Evaluation of drug sensitivity, immunological characteristics, and prognosis in melanoma patients using an endoplasmic reticulum stress-associated signature based on bioinformatics and pan-cancer analysis. J Mol Med (Berl) 2023; 101:1267-1287. [PMID: 37653150 DOI: 10.1007/s00109-023-02365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 05/27/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023]
Abstract
We aimed to develop endoplasmic reticulum (ER) stress-related risk signature to predict the prognosis of melanoma and elucidate the immune characteristics and benefit of immunotherapy in ER-related risk score-defined subgroups of melanoma based on a machine learning algorithm. Based on The Cancer Genome Atlas (TCGA) melanoma dataset (n = 471) and GTEx database (n = 813), 365 differentially expressed ER-associated genes were selected using the univariate Cox model and LASSO penalty Cox model. Ten genes impacting OS were identified to construct an ER-related signature by using the multivariate Cox regression method and validated with the Gene Expression Omnibus (GEO) dataset. Thereafter, the immune features, CNV, methylation, drug sensitivity, and the clinical benefit of anticancer immune checkpoint inhibitor (ICI) therapy in risk score subgroups, were analyzed. We further validated the gene signature using pan-cancer analysis by comparing it to other tumor types. The ER-related risk score was constructed based on the ARNTL, AGO1, TXN, SORL1, CHD7, EGFR, KIT, HLA-DRB1 KCNA2, and EDNRB genes. The high ER stress-related risk score group patients had a poorer overall survival (OS) than the low-risk score group patients, consistent with the results in the GEO cohort. The combined results suggested that a high ER stress-related risk score was associated with cell adhesion, gamma phagocytosis, cation transport, cell surface cell adhesion, KRAS signalling, CD4 T cells, M1 macrophages, naive B cells, natural killer (NK) cells, and eosinophils and less benefitted from ICI therapy. Based on the expression patterns of ER stress-related genes, we created an appropriate predictive model, which can also help distinguish the immune characteristics, CNV, methylation, and the clinical benefit of ICI therapy. KEY MESSAGES: Melanoma is the cutaneous tumor with a high degree of malignancy, the highest fatality rate, and extremely poor prognosis. Model usefulness should be considered when using models that contained more features. We constructed the Endoplasmic Reticulum stress-associated signature using TCGA and GEO database based on machine learning algorithm. ER stress-associated signature has excellent ability for predicting prognosis for melanoma.
Collapse
Affiliation(s)
| | - Bingqian Hu
- Department of Dermatology, Xiangya Hospital of Central South University, Changsha, 410000, China
| | - Zheng Wang
- School of Computer Science, Hunan First Normal University, Changsha, 410205, China
| | - Jiaoju Wang
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Cong Cao
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Jianglin Zhang
- Department of Dermatology, The Second Clinical Medical College, Shenzhen People's Hospital Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Muzhou Hou
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China.
| | - Min Qi
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
7
|
Xia B, Li Q, Zheng K, Wu J, Huang C, Liu K, You Q, Yuan X. Down-regulation of Hrd1 protects against myocardial ischemia-reperfusion injury by regulating PPARα to prevent oxidative stress, endoplasmic reticulum stress, and cellular apoptosis. Eur J Pharmacol 2023; 954:175864. [PMID: 37392829 DOI: 10.1016/j.ejphar.2023.175864] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 07/03/2023]
Abstract
The E3 ubiquitin ligase HMG-CoA reductase degradation protein 1 (Hrd1) is a key enzyme for ER-associated degradation of misfolded proteins. Its role in ischemic heart disease has not been fully elucidated. Here, we investigated its effect on oxidative status and cell survival in cardiac ischemia-reperfusion injury (MIRI). We found that virus-induced down-regulation of Hrd1 expression limited infarct size, decreased creatinine kinase (CK) and lactate dehydrogenase (LDH), and preserved cardiac function in mice subjected to left anterior descending coronary artery ligation and reperfusion. Silencing of the Hrd1 gene also prevented the ischemia/reperfusion (I/R)-induced (i) increase in dihydroethidium (DHE) intensity, mitochondrial production of reactive oxygen species (ROS), malondialdehyde (MDA), and nitric oxide (NO), (ii) decrease in total antioxidant capacity (T-AOC) and glutathione (GSH), (iii) disruption of mitochondrial membrane potential, and (iv) increase in the expression of glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) in ischemic heart tissue. In addition, down-regulation of Hrd1 expression prevented the abnormally increased caspase-3/caspase-9/Bax expression and decreased Bcl-2 expression in ischemic heart tissue of I/R mice. Further analysis showed that the I/R stimulus reduced peroxisome proliferation activated receptor α (PPARα) expression in ischemic heart tissue, which was partially prevented by down-regulation of Hrd1. Pharmacological inhibition of PPARα was able to abolish the preventive effect of down-regulation of Hrd1 on oxidative stress, endoplasmic reticulum stress, and cellular apoptosis in ischemic heart tissue. These data suggest that down-regulation of Hrd1 protects the heart from I/R-induced damage by suppressing oxidative stress and cellular apoptosis likely through PPARα.
Collapse
Affiliation(s)
- Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Koulong Zheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Miao J, Yao S, Sun H, Jiang Z, Gao Z, Xu J, Chen K. Protective Effect of Water-Soluble Acacetin Prodrug on APAP-Induced Acute Liver Injury Is Associated with Upregulation of PPARγ and Alleviation of ER Stress. Int J Mol Sci 2023; 24:11320. [PMID: 37511082 PMCID: PMC10380069 DOI: 10.3390/ijms241411320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
A water-soluble acacetin prodrug has been synthesized and reported by our group previously. Acetaminophen (APAP) overdose is a leading cause of acute liver injury. We found that subcutaneous injection of acacetin prodrug (5, 10, 20 mg/kg) decreased serum ALT, AST, and ALP, corrected the abnormal MDA and GSH in liver, and improved intrahepatic hemorrhage and destruction of liver structures in APAP (300 mg/kg)-treated mice. Molecular mechanism analysis revealed that the expressions of endoplasmic reticulum (ER) stress markers ATF6, CHOP, and p-PERK, apoptosis-related protein BAX, and cleaved caspase 3 were decreased by acacetin in a dose-dependent manner in vivo and in vitro. Moreover, via the acacetin-upregulated peroxisome-proliferator-activated receptor gamma (PPARγ) of HepG2 cells and liver, the suppressive effect of acacetin on ER stress and apoptosis was abolished by PPARγ inhibitor (GW9662) or PPARγ-siRNA. Molecular docking revealed that acacetin can bind to three active pockets of PPARγ, mainly by hydrogen bond. Our results provide novel evidence that acacetin prodrug exhibits significant protective effect against APAP-induced liver injury by targeting PPARγ, thereby suppressing ER stress and hepatocyte apoptosis. Acacetin prodrug is likely a promising new drug candidate for treating patients with acute liver injury induced by APAP.
Collapse
Affiliation(s)
- Jiaen Miao
- Department of Pharmacology, Ningbo University School of Medicine, 818 Fenghua Rd., Ningbo 315100, China
| | - Shujun Yao
- Department of Pharmacology, Ningbo University School of Medicine, 818 Fenghua Rd., Ningbo 315100, China
| | - Hao Sun
- Department of Pharmacology, Ningbo University School of Medicine, 818 Fenghua Rd., Ningbo 315100, China
| | - Zhe Jiang
- Department of Pharmacology, Ningbo University School of Medicine, 818 Fenghua Rd., Ningbo 315100, China
| | - Zhe Gao
- The Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo 315000, China
| | - Jia Xu
- Department of Pharmacology, Ningbo University School of Medicine, 818 Fenghua Rd., Ningbo 315100, China
| | - Kuihao Chen
- Department of Pharmacology, Ningbo University School of Medicine, 818 Fenghua Rd., Ningbo 315100, China
| |
Collapse
|
9
|
Kim TW. Nodakenin Induces ROS-Dependent Apoptotic Cell Death and ER Stress in Radioresistant Breast Cancer. Antioxidants (Basel) 2023; 12:antiox12020492. [PMID: 36830050 PMCID: PMC9952086 DOI: 10.3390/antiox12020492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Angelica gigas exerts powerful anti-tumor and anti-cancer effects in various cancer cell types. However, there have been few studies regarding the anti-cancer effect of nodakenin, a bioactive compound of Angelica gigas, in vivo and in vitro on breast cancers. I found that nodakenin, in a concentration-dependent manner, inhibits breast cancer cell viability and decreases the tumor volume in mice. Additionally, nodakenin induces caspase-3-dependent apoptosis in breast cancer cells; however, the combination of Z-VAD-FMK and nodakenin suppresses the caspase-3-dependent apoptotic cell death. Furthermore, nodakenin mediates apoptotic cell death via the PERK-mediated signaling pathway and calcium (Ca2+) release, and nodakenin combined with thapsigargin induces synergistic cell death by inhibiting sarco/endoplasmic reticulum (ER) Ca2+-ATPase. However, knockdown of PERK or CHOP inhibits Ca2+ generation and caspase-dependent apoptosis in nodakenin-treated breast cancer cells. Nodakenin induces ROS and Ca2+ generation, ER stress, and apoptotic cell death; however, the knockdown of Nox4 inhibits ROS generation and ER stress- and caspase-dependent apoptotic cell death. In addition, nodakenin combined with radiation overcomes radioresistance in radioresistant breast cancer cells by suppressing epithelial-mesenchymal transition phenotypes, including the decrease in E-cadherin and the increase in N-cadherin and vimentin. Therefore, these findings indicate that nodakenin may be a novel therapeutic strategy for breast cancers.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, 123 Dongdae-ro, Gyeongju 38066, Gyeongbuk, Republic of Korea
| |
Collapse
|
10
|
Kwak AW, Lee JY, Lee SO, Seo JH, Park JW, Choi YH, Cho SS, Yoon G, Lee MH, Shim JH. Echinatin induces reactive oxygen species-mediated apoptosis via JNK/p38 MAPK signaling pathway in colorectal cancer cells. Phytother Res 2023; 37:563-577. [PMID: 36184899 DOI: 10.1002/ptr.7634] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/15/2022] [Accepted: 09/03/2022] [Indexed: 11/05/2022]
Abstract
Colorectal cancer (CRC) is a very common and deadly cancer worldwide, and oxaliplatin is used as first-line chemotherapy. However, resistance usually develops, limiting treatment. Echinatin (Ech) is the main component of licorice and exhibits various therapeutic effects on inflammation-mediated diseases and cancer, ischemia/reperfusion, and liver injuries. The present study elucidated the underlying molecular mechanism of Ech-induced apoptosis in both oxaliplatin-sensitive (HT116 and HT29) and -resistant (HCT116-OxR and HT29-OxR) CRC cells. To evaluate the antiproliferative activities of Ech, we performed MTT and soft agar assays. Ech reduced viability, colony size, and numbers of CRC cells. The underlying molecular mechanisms were explored by various flow cytometry analyses. Ech-induced annexin-V stained cells, reactive oxygen species (ROS) generation, cell cycle arrest, JNK/p38 MAPK activation, endoplasmic reticulum (ER) stress, mitochondrial membrane potential depolarization, and multi-caspase activity. In addition apoptosis-, cell cycle-, and ER stress-related protein levels were confirmed by western blotting. Moreover, we verified ROS-mediated cell death by treatment with inhibitors such as N-acetyl-L-cysteine, SP600125, and SB203580. Taken together, Ech exhibits anticancer activity in oxaliplatin-sensitive and -resistant CRCs by inducing ROS-mediated apoptosis through the JNK/p38 MAPK signaling pathway. This is the first study to show that Ech has the potential to treat drug-resistant CRC, providing new directions for therapeutic strategies targeting drug-resistant CRC.
Collapse
Affiliation(s)
- Ah-Won Kwak
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Republic of Korea
| | - Jin-Young Lee
- Department of Biological Sciences, Keimyung University, Daegu, Republic of Korea
| | - Seung-On Lee
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Cheonggye-myeon, Jeonnam, Republic of Korea
| | - Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jin Woo Park
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Republic of Korea.,Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Cheonggye-myeon, Jeonnam, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Republic of Korea.,Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Cheonggye-myeon, Jeonnam, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Republic of Korea
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan, Republic of Korea.,Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, College of Pharmacy, Mokpo National University, Cheonggye-myeon, Jeonnam, Republic of Korea.,The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
11
|
Yin H, Liu Y, Yue H, Tian Y, Dong P, Xue C, Zhao YT, Zhao Z, Wang J. DHA- and EPA-Enriched Phosphatidylcholine Suppress Human Lung Carcinoma 95D Cells Metastasis via Activating the Peroxisome Proliferator-Activated Receptor γ. Nutrients 2022; 14:nu14214675. [PMID: 36364935 PMCID: PMC9654432 DOI: 10.3390/nu14214675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
The antineoplastic effects of docosahexaenoic acid-containing phosphatidylcholine (DHA-PC) and eicosapentaenoic acid-containing phosphatidylcholine (EPA-PC) were explored, and their underlying mechanisms in the human lung carcinoma 95D cells (95D cells) were investigated. After treatment of 95D cells with DHA-PC or EPA-PC, cell biological behaviors such as growth, adhesion, migration, and invasion were studied. Immunofluorescence and western blotting were carried out to assess underlying molecular mechanisms. Results showed that 95D cells proliferation and adherence in the DHA-PC or EPA-PC group were drastically inhibited than the control group. DHA-PC and EPA-PC suppressed the migration and invasion of 95D cells by disrupting intracellular F-actin, which drives cell movement. The protein expression of PPARγ was induced versus the control group. Furthermore, critical factors related to invasion, including matrix metallopeptidase 9 (MMP9), heparanase (Hpa), and vascular endothelial growth factor (VEGF), were drastically downregulated through the PPARγ/NF-κB signaling pathway. C-X-C chemokine receptor type 4 (CXCR4) and cofilin were significantly suppressed via DHA-PC and EPA-PC through the PPARγ/phosphatase and tensin homolog (PTEN)/serine-threonine protein kinase (AKT) signaling pathway. DHA-PC and EPA-PC reversed the PPARγ antagonist GW9662-induced reduction of 95D cells in migration and invasion capacity, suggesting that PPARγ was directly involved in the anti-metastasis efficacy of DHA-PC and EPA-PC. In conclusion, DHA-PC and EPA-PC have great potential for cancer therapy, and the antineoplastic effects involve the activation of PPARγ. EPA-PC showed more pronounced antineoplastic effects than DHA-PC, possibly due to the more robust activation of PPARγ by EPA-PC.
Collapse
Affiliation(s)
- Haowen Yin
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Yuanyuan Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Hao Yue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Yingying Tian
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Ping Dong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Yun-Tao Zhao
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zifang Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Hainan Huayan Collagen Technology Co., Ltd., Haikou 571000, China
- Correspondence: (Z.Z.); (J.W.); Tel.: +86-898-6655-3777 (Z.Z.); +86-532-8203-1967 (J.W.)
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
- Correspondence: (Z.Z.); (J.W.); Tel.: +86-898-6655-3777 (Z.Z.); +86-532-8203-1967 (J.W.)
| |
Collapse
|
12
|
Targeting Nuclear Receptors in Lung Cancer—Novel Therapeutic Prospects. Pharmaceuticals (Basel) 2022; 15:ph15050624. [PMID: 35631448 PMCID: PMC9145966 DOI: 10.3390/ph15050624] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023] Open
Abstract
Lung cancer, the second most commonly diagnosed cancer, is the major cause of fatalities worldwide for both men and women, with an estimated 2.2 million new incidences and 1.8 million deaths, according to GLOBOCAN 2020. Although various risk factors for lung cancer pathogenesis have been reported, controlling smoking alone has a significant value as a preventive measure. In spite of decades of extensive research, mechanistic cues and targets need to be profoundly explored to develop potential diagnostics, treatments, and reliable therapies for this disease. Nuclear receptors (NRs) function as transcription factors that control diverse biological processes such as cell growth, differentiation, development, and metabolism. The aberrant expression of NRs has been involved in a variety of disorders, including cancer. Deregulation of distinct NRs in lung cancer has been associated with numerous events, including mutations, epigenetic modifications, and different signaling cascades. Substantial efforts have been made to develop several small molecules as agonists or antagonists directed to target specific NRs for inhibiting tumor cell growth, migration, and invasion and inducing apoptosis in lung cancer, which makes NRs promising candidates for reliable lung cancer therapeutics. The current work focuses on the importance of various NRs in the development and progression of lung cancer and highlights the different small molecules (e.g., agonist or antagonist) that influence NR expression, with the goal of establishing them as viable therapeutics to combat lung cancer.
Collapse
|
13
|
Crassolide Induces G2/M Cell Cycle Arrest, Apoptosis, and Autophagy in Human Lung Cancer Cells via ROS-Mediated ER Stress Pathways. Int J Mol Sci 2022; 23:ijms23105624. [PMID: 35628435 PMCID: PMC9144222 DOI: 10.3390/ijms23105624] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/05/2022] [Accepted: 05/15/2022] [Indexed: 02/07/2023] Open
Abstract
Crassolide, a cembranoid diterpene extracted from the soft coral Lobophytum crissum, has been proven to possess antioxidant and immunomodulatory properties. In the present study, we assessed the anticancer effects of crassolide on human H460 non-small-cell lung cancer (NSCLC) cells. We found that crassolide exerted cytotoxic effects on H460 cancer cells in vitro, inducing G2/M phase arrest and apoptosis. In addition, in H460 cells exposed to crassolide, the expression of the autophagy-related proteins LC3-II and beclin was increased, while the expression of p62 was decreased. Moreover, inhibiting autophagy with chloroquine (CQ) suppressed the crassolide-induced G2/M arrest and apoptosis of H460 cells. Moreover, we also found that crassolide induced endoplasmic reticulum (ER) stress in lung cancer cells by increasing the expression of ER stress marker proteins and that the crassolide-induced G2/M arrest, apoptosis, and autophagy were markedly attenuated by the ER stress inhibitor 4-phenylbutyric acid (4-PBA). Furthermore, we found that crassolide promoted reactive oxygen species (ROS) production by H460 cells and that the ROS inhibitor N-acetylcysteine (NAC) decreased the crassolide-induced ER stress, G2/M arrest, apoptosis, and autophagy. In conclusion, our findings show that crassolide inhibits NSCLC cell malignant biological behaviors for the first time, suggesting that this effect may be mechanistically achieved by inducing G2/M arrest, apoptosis, and autophagy through ROS accumulation, which activates the ER stress pathway. As a result of our findings, we now have a better understanding of the molecular mechanism underlying the anticancer effect of crassolide, and we believe crassolide might be a candidate for targeted cancer therapy.
Collapse
|
14
|
Lei S, Zhang J, Blum NT, Li M, Zhang DY, Yin W, Zhao F, Lin J, Huang P. In vivo three-dimensional multispectral photoacoustic imaging of dual enzyme-driven cyclic cascade reaction for tumor catalytic therapy. Nat Commun 2022; 13:1298. [PMID: 35277519 PMCID: PMC8917194 DOI: 10.1038/s41467-022-29082-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 02/24/2022] [Indexed: 01/08/2023] Open
Abstract
Non-invasive visualization of dynamic molecular events in real-time via molecular imaging may enable the monitoring of cascade catalytic reactions in living systems, however effective imaging modalities and a robust catalytic reaction system are lacking. Here we utilize three-dimensional (3D) multispectral photoacoustic (PA) molecular imaging to monitor in vivo cascade catalytic therapy based on a dual enzyme-driven cyclic reaction platform. The system consists of a two-dimensional (2D) Pd-based nanozyme conjugated with glucose oxidase (GOx). The combination of nanozyme and GOx can induce the PA signal variation of endogenous molecules. Combined with the PA response of the nanozyme, we can simultaneously map the 3D PA signals of dynamic endogenous and exogenous molecules associated with the catalytic process, thus providing a real-time non-invasive visualization. We can also treat tumors under the navigation of the PA imaging. Therefore, our study demonstrates the imaging-guided potential of 3D multispectral PA imaging in feedback-looped cascade catalytic therapy.
Collapse
Affiliation(s)
- Shan Lei
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jing Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Nicholas Thomas Blum
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Meng Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Dong-Yang Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Weimin Yin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Feng Zhao
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
15
|
Fu L, Jin W, Zhang J, Zhu L, Lu J, Zhen Y, Zhang L, Ouyang L, Liu B, Yu H. Repurposing non-oncology small-molecule drugs to improve cancer therapy: Current situation and future directions. Acta Pharm Sin B 2022; 12:532-557. [PMID: 35256933 PMCID: PMC8897051 DOI: 10.1016/j.apsb.2021.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/05/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022] Open
Abstract
Drug repurposing or repositioning has been well-known to refer to the therapeutic applications of a drug for another indication other than it was originally approved for. Repurposing non-oncology small-molecule drugs has been increasingly becoming an attractive approach to improve cancer therapy, with potentially lower overall costs and shorter timelines. Several non-oncology drugs approved by FDA have been recently reported to treat different types of human cancers, with the aid of some new emerging technologies, such as omics sequencing and artificial intelligence to overcome the bottleneck of drug repurposing. Therefore, in this review, we focus on summarizing the therapeutic potential of non-oncology drugs, including cardiovascular drugs, microbiological drugs, small-molecule antibiotics, anti-viral drugs, anti-inflammatory drugs, anti-neurodegenerative drugs, antipsychotic drugs, antidepressants, and other drugs in human cancers. We also discuss their novel potential targets and relevant signaling pathways of these old non-oncology drugs in cancer therapies. Taken together, these inspiring findings will shed new light on repurposing more non-oncology small-molecule drugs with their intricate molecular mechanisms for future cancer drug discovery.
Collapse
|
16
|
Lee SO, Joo SH, Kwak AW, Lee MH, Seo JH, Cho SS, Yoon G, Chae JI, Shim JH. Podophyllotoxin Induces ROS-Mediated Apoptosis and Cell Cycle Arrest in Human Colorectal Cancer Cells via p38 MAPK Signaling. Biomol Ther (Seoul) 2021; 29:658-666. [PMID: 34642263 PMCID: PMC8551740 DOI: 10.4062/biomolther.2021.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/29/2022] Open
Abstract
Podophyllotoxin (PT), a lignan compound from the roots and rhizomes of Podophyllum peltatum, has diverse pharmacological activities including anticancer effect in several types of cancer. The molecular mechanism of the anticancer effects of PT on colorectal cancer cells has not been reported yet. In this study, we sought to evaluate the anticancer effect of PT on human colorectal cancer HCT116 cells and identify the detailed molecular mechanism. PT inhibited the growth of cells and colony formation in a concentration-dependent manner and induced apoptosis as determined by the annexin V/7-aminoactinomycin D double staining assay. PT-induced apoptosis was accompanied by cell cycle arrest in the G2/M phase and an increase in the generation of reactive oxygen species (ROS). The effects of PT on the induction of ROS and apoptosis were prevented by pretreatment with N-acetyl-L-cysteine (NAC), indicating that an increase in ROS generation mediates the apoptosis of HCT116 cells induced by PT. Furthermore, Western blot analysis showed that PT upregulated the level of phospho (p)-p38 mitogen-activated protein kinase (MAPK). The treatment of SB203580, a p38 inhibitor, strongly prevented the apoptosis induced by PT, suggesting that PT-induced apoptosis involved the p38 MAPK signaling pathway. In addition, PT induced the loss of mitochondrial membrane potential and multi-caspase activation. The results suggested that PT induced cell cycle arrest in the G2/M phase and apoptosis through the p38 MAPK signaling pathway by upregulating ROS in HCT116 cells.
Collapse
Affiliation(s)
- Seung-On Lee
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Republic of Korea
| | - Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Ah-Won Kwak
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea
| | - Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Republic of Korea.,Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea.,The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| |
Collapse
|
17
|
Muzio G, Barrera G, Pizzimenti S. Peroxisome Proliferator-Activated Receptors (PPARs) and Oxidative Stress in Physiological Conditions and in Cancer. Antioxidants (Basel) 2021; 10:antiox10111734. [PMID: 34829605 PMCID: PMC8614822 DOI: 10.3390/antiox10111734] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor superfamily. Originally described as “orphan nuclear receptors”, they can bind both natural and synthetic ligands acting as agonists or antagonists. In humans three subtypes, PPARα, β/δ, γ, are encoded by different genes, show tissue-specific expression patterns, and contribute to the regulation of lipid and carbohydrate metabolisms, of different cell functions, including proliferation, death, differentiation, and of processes, as inflammation, angiogenesis, immune response. The PPAR ability in increasing the expression of various antioxidant genes and decreasing the synthesis of pro-inflammatory mediators, makes them be considered among the most important regulators of the cellular response to oxidative stress conditions. Based on the multiplicity of physiological effects, PPAR involvement in cancer development and progression has attracted great scientific interest with the aim to describe changes occurring in their expression in cancer cells, and to investigate the correlation with some characteristics of cancer phenotype, including increased proliferation, decreased susceptibility to apoptosis, malignancy degree and onset of resistance to anticancer drugs. This review focuses on mechanisms underlying the antioxidant and anti-inflammatory properties of PPARs in physiological conditions, and on the reported beneficial effects of PPAR activation in cancer.
Collapse
|
18
|
Gong TT, Guo Q, Li X, Zhang TN, Liu FH, He XH, Lin B, Wu QJ. Isothiocyanate Iberin inhibits cell proliferation and induces cell apoptosis in the progression of ovarian cancer by mediating ROS accumulation and GPX1 expression. Biomed Pharmacother 2021; 142:111533. [PMID: 34148735 DOI: 10.1016/j.biopha.2021.111533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/12/2021] [Accepted: 03/21/2021] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecologic malignancies with poor survival rate, and Iberin is a member of isothiocyanate family with anti-tumor activity. However, the role of Iberin in OC development has not been reported yet. In this study, A2780 and OVCAR-3 cells were treated with gradient concentrations of Iberin to investigate the effect of Iberin on OC in vitro. Meanwhile, the in vivo tumorgenesis experiment was performed using female BALB/c nude mice treated with Iberin. Iberin inhibited cell proliferation, induced G2 cell cycle arrest and promoted cell apoptosis in OC cells. Besides, Iberin reduced GSH/GSSG level, enhanced ROS accumulation, and activated MAPK signaling in OC cells. More interestingly, ROS scavenger (NAC) compensated the anti-proliferative and pro-apoptotic effects of Iberin on OC cells, suggesting the involvement of ROS in the regulation of Iberin on OC cell growth. Notably, Iberin induced down-regulation of glutathione peroxidase-1 (GPX1), and over-expression of GPX1 reversed Iberin-mediated alterations in the proliferation, apoptosis and ROS accumulation of OC cells. The in vivo tumorgenesis study further evidenced the protection of Iberin against OC development. Besides, Iberin displayed a synergistic effect on the enhancement of chemo-sensitivity in OC cells. In summary, our study demonstrates the anti-tumor effect of Iberin on OC and its potential as a therapeutic agent against OC in the future.
Collapse
Affiliation(s)
- Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Qian Guo
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xin-Hui He
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| |
Collapse
|
19
|
Zhang W, Wang W, Xu M, Xie H, Pu Z. GPR43 regulation of mitochondrial damage to alleviate inflammatory reaction in sepsis. Aging (Albany NY) 2021; 13:22588-22610. [PMID: 34584017 PMCID: PMC8507289 DOI: 10.18632/aging.203572] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022]
Abstract
Sepsis is a common critical illness in ICU and always a great difficulty in clinical treatment. GPR43 (G protein-coupled receptor 43) participates in regulating appetite and gastrointestinal peptide secretion to modulate fat decomposition and formation. However, the biological contribution of GPR43 on inflammation of sepsis has not been previously investigated. We investigated the mechanisms of GPR43 gene, which plays a possible role in distinguishing sepsis and contributes to the pathogenesis of sepsis-induced inflammatory reaction. Furthermore, we performed studies with mice induced to sepsis by Cecal Ligation and Puncture (CLP), Knockout GPR43 (GPR43-/-) mice, and Wild Type (WT) mice induced with CLP. In addition, lung tissues and cell samples were analyzed by histology, Quantitative Polymerase Chain Reaction (Q-PCR), Enzyme-linked Immunosorbent (ELISA) Assay, and western blot. GPR43 agonist could significantly reduce inflammation reactions and trigger lung injury in mice with sepsis. As for GPR43-/- mice, the risks of sepsis-induced inflammatory reactions and corresponding lung injury were promoted. On the one hand, the up-regulation of GPR43 gene reduced ROS mitochondrial damage to inhibit inflammatory reactions via the inactivation of NLRP3 Inflammasome by PPARγ/ Nox1/EBP50/ p47phox signal channel. On the other hand, the down-regulation of GPR43 promoted inflammatory reactions in vitro model through the acceleration of ROS-dependently mitochondrial damage by PPARγ/ Nox1/EBP50/ p47phox/ NLRP3 signal channel. These findings indicate that the inhibition of GPR43 as a possible important factor of sepsis may shed lights on the mechanism of sepsis-induced inflammation reaction.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Pharmacy, Second Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Wusan Wang
- Department of Pharmacology, College of Pharmacy, Wannan Medical College, Wuhu 241002, Anhui, China
| | - Maodi Xu
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Haitang Xie
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Zhichen Pu
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China.,State Key Laboratory of Natural Medicines, Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
20
|
Cheng HS, Yip YS, Lim EKY, Wahli W, Tan NS. PPARs and Tumor Microenvironment: The Emerging Roles of the Metabolic Master Regulators in Tumor Stromal-Epithelial Crosstalk and Carcinogenesis. Cancers (Basel) 2021; 13:2153. [PMID: 33946986 PMCID: PMC8125182 DOI: 10.3390/cancers13092153] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been extensively studied for more than three decades. Consisting of three isotypes, PPARα, γ, and β/δ, these nuclear receptors are regarded as the master metabolic regulators which govern many aspects of the body energy homeostasis and cell fate. Their roles in malignancy are also increasingly recognized. With the growing interest in crosstalk between tumor stroma and epithelium, this review aims to highlight the current knowledge on the implications of PPARs in the tumor microenvironment. PPARγ plays a crucial role in the metabolic reprogramming of cancer-associated fibroblasts and adipocytes, coercing the two stromal cells to become substrate donors for cancer growth. Fibroblast PPARβ/δ can modify the risk of tumor initiation and cancer susceptibility. In endothelial cells, PPARβ/δ and PPARα are pro- and anti-angiogenic, respectively. Although the angiogenic role of PPARγ remains ambiguous, it is a crucial regulator in autocrine and paracrine signaling of cancer-associated fibroblasts and tumor-associated macrophages/immune cells. Of note, angiopoietin-like 4 (ANGPTL4), a secretory protein encoded by a target gene of PPARs, triggers critical oncogenic processes such as inflammatory signaling, extracellular matrix derangement, anoikis resistance and metastasis, making it a potential drug target for cancer treatment. To conclude, PPARs in the tumor microenvironment exhibit oncogenic activities which are highly controversial and dependent on many factors such as stromal cell types, cancer types, and oncogenesis stages. Thus, the success of PPAR-based anticancer treatment potentially relies on innovative strategies to modulate PPAR activity in a cell type-specific manner.
Collapse
Affiliation(s)
- Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
| | - Yun Sheng Yip
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
| | - Eldeen Kai Yi Lim
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore;
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP-PURPAN, UMR 1331, UPS, Université de Toulouse, 31300 Toulouse, France
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore; (Y.S.Y.); (W.W.)
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore;
| |
Collapse
|
21
|
The Novel Benzothiazole Derivative PB11 Induces Apoptosis via the PI3K/AKT Signaling Pathway in Human Cancer Cell Lines. Int J Mol Sci 2021; 22:ijms22052718. [PMID: 33800261 PMCID: PMC7962528 DOI: 10.3390/ijms22052718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/23/2021] [Accepted: 03/04/2021] [Indexed: 11/17/2022] Open
Abstract
Among several anti-cancer therapies, chemotherapy can be used regardless of the stage of the disease. However, development of anti-cancer agents from potential chemicals must be executed very cautiously because of several problems, such as safety, drug resistance, and continuous administration. Most chemotherapeutics selectively cause cancer cells to undergo apoptosis. In this study, we tested the effects of a novel chemical, the benzothiazole derivative N-[2-[(3,5-dimethyl-1,2-oxazol-4-yl)methylsulfanyl]-1,3-benzothiazol-6-yl]-4-oxocyclohexane-1-carboxamide (PB11) on the human cell lines U87 (glioblastoma), and HeLa (cervix cancer). It was observed that this chemical was highly cytotoxic for these cells (IC50s < 50 nM). In addition, even 40 nM PB11 induced the classical apoptotic symptoms of DNA fragmentation and nuclear condensation. The increase of caspase-3 and -9 activities also indicated an increased rate of apoptosis, which was further confirmed via Western blotting analysis of apoptosis-associated proteins. Accordingly, PB11 treatment up-regulated the cellular levels of caspase-3 and cytochrome-c, whereas it down-regulated PI3K and AKT. These results suggest that PB11 induces cytotoxicity and apoptosis in cancer cells by suppressing the PI3K/AKT signaling pathways and, thus, may serve as an anti-cancer therapeutic.
Collapse
|