1
|
CHOI HYEONGSIM, KU JEONGKUI, KO SEONGGYU, YUN PILYOUNG. Anticancer effects of SH003 and its active component Cucurbitacin D on oral cancer cell lines via modulation of EMT and cell viability. Oncol Res 2025; 33:1217-1227. [PMID: 40296905 PMCID: PMC12034017 DOI: 10.32604/or.2025.059791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/03/2025] [Indexed: 04/30/2025] Open
Abstract
Background Oral cancer remains a significant global health challenge, as it has high morbidity and mortality rates. Current treatments show limited efficacy and have severe side effects, prompting searches for new therapeutic agents. SH003, a traditional herbal formulation comprising Astragalus membranaceus, Angelica gigas, and Trichosanthes kirilowii, has demonstrated potential anticancer properties in previous studies. However, its specific efficacy against oral cancer and the role of its key components, particularly Cucurbitacin D, remain underexplored. Methods The cytotoxic effects of SH003 and its major components-i.e., Cucurbitacin D, Decursin, Formononetin, and Nodakenin-were evaluated using 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT), Trypan Blue exclusion, and Lactate Dehydrogenase (LDH) release assays. Cell migration was analyzed via wound healing assays, and apoptosis induction was assessed using cell cycle analysis and caspase activation assays. Epithelial-to-mesenchymal transition (EMT) marker expression (E-cadherin and N-cadherin) was measured using Western blotting and Quantitative reverse transcription PCR (qRT-PCR). Results SH003 significantly reduced cell viability in a dose-dependent manner, with YD-8 and YD-9 cells showing greater sensitivity than YD-38 cells. Of the individual compounds, Cucurbitacin D was identified as a key active agent, as it exhibited potent inhibition of cell migration and significant modulation of EMT markers, including the upregulation of E-cadherin and downregulation of N-cadherin. These effects were most pronounced in YD-9 cells. Conclusions Taken together, these findings suggest that Cucurbitacin D plays a crucial role mediating the anticancer activity of SH003, particularly via the reversal of EMT and the reduction of migratory and invasive potential of oral cancer cells. This study provides valuable insight into the mechanistic basis of SH003, highlighting its potential as a therapeutic agent against oral cancer. Further research, including in vivo studies and clinical trials, is needed to elucidate its precise mechanisms and potential applications against other cancer types.
Collapse
Affiliation(s)
- HYEONG SIM CHOI
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - JEONG-KUI KU
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - SEONG-GYU KO
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02435, Republic of Korea
| | - PIL-YOUNG YUN
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
- Department of Dentistry and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| |
Collapse
|
2
|
Chuang PC, Su WH, Hsieh CH, Huang EY. TIAM2S Operates Multifaced Talents to Alleviate Radiosensitivity, Restrict Apoptosis, Provoke Cell Propagation, and Escalate Cell Migration for Aggravating Radioresistance-Intensified Cervical Cancer Progression. Cells 2025; 14:339. [PMID: 40072068 PMCID: PMC11898548 DOI: 10.3390/cells14050339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Radioresistance remains a major obstacle in cervical cancer treatment, frequently engendering tumor relapse and metastasis. However, the details of its mechanism of action remain largely enigmatic. This study delineates the prospective impacts of short-form human T-cell lymphoma invasion and metastasis 2 (TIAM2S) involving the radiation resistance of cervical cancer. In this study, we established three pairs of radioresistant (RR) cervical cancer cells (HeLa, C33A and CaSki) and their parental wild-type (WT) cells. We revealed a consistent augmentation of TIAM2S, but not long-form human T-cell lymphoma invasion and metastasis 2 (TIAM2L) were displayed in RR cells that underwent a 6 Gy radiation administration. Remarkably, RR cells exhibited decreased radiosensitivity and abridged apoptosis, as estimated through a clonogenic survival curve assay and Annexin V/Propidium Iodide apoptosis assay, respectively. TIAM2S suppression increased radiosensitivity and enhanced cell apoptosis in RR cells, whereas its forced introduction modestly abolished radiosensitivity and diminished WT cell apoptosis. Furthermore, TIAM2S overexpression notably aggravated RR cell migration, whereas its blockage reduced WT cell mobilities, as confirmed by an in vitro time-lapse recording assay. Notably, augmented lung localization was revealed after a tail-vein injection of CaSki-RR cells using the in vivo short-term lung locomotion BALB/c nude mouse model. TIAM2S impediment notably reduced radioresistance-increased lung locomotion. This study provides evidence that TIAM2S may operate as an innovative signature in cervical cancer that is resistant to radiotherapy. It displays multi-faceted roles including radioprotection, restricting apoptosis, promoting cell proliferation, and escalating cell migration/metastasis. Targeting TIAM2S, together with conventional radiotherapy, may be an innovative strategy for intensifying radiosensitivity and protecting against subsequent uncontrolled tumor growth and metastasis in cervical cancer treatment.
Collapse
Affiliation(s)
- Pei-Chin Chuang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (P.-C.C.); (W.-H.S.)
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807017, Taiwan
| | - Wen-Hong Su
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (P.-C.C.); (W.-H.S.)
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Eng-Yen Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| |
Collapse
|
3
|
Liang C, Liu X, Yu J, Shi L, Wei W, Zhu Y, Feng M, Tang T, Li D, Yang T, Zheng J, Ma B, Wei L. Hypericin photoactivation induces triple-negative breast cancer cells pyroptosis by targeting the ROS/CALR/Caspase-3/GSDME pathway. J Adv Res 2025:S2090-1232(25)00059-1. [PMID: 39870303 DOI: 10.1016/j.jare.2025.01.042] [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: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025] Open
Abstract
INTRODUCTION Hypericin (HP), a natural photosensitizer, has demonstrated great efficacy in photodynamic therapy (PDT) for cancer treatment. In addition to the induction of apoptosis and necrosis through reactive oxygen species (ROS) generation, the therapeutic mechanisms and targets of PDT-HP remain unknown. OBJECTIVES To investigate the direct targets and mechanisms of action of photoactivated hypericin in the inhibition of triple-negative breast cancer (TNBC). METHODS Cell pyroptosis was examined via LDH release, SYTOX Green staining, and ELISA. RNA sequencing, network pharmacology, drug affinity target stability (DARTS)-tandem mass spectrometry (MS/MS), and molecular docking were employed to identify drug targets. Furthermore, immunoblotting and flow cytometry were utilized to elucidate the mechanisms of drug action. RESULTS Our research revealed that PDT-HP can induce pyroptosis in TNBC cells. Further investigation revealed that PDT-HP induces endoplasmic reticulum stress, activating Caspase-3 and gasdermin E (GSDME) to trigger TNBC cell pyroptosis. RNA-seq, network pharmacology, and DARTS-MS/MS proteomic analyses revealed that the endoplasmic reticulum protein calreticulin (CALR) is a potential HP target and that interfering with CALR inhibited PDT-HP-induced pyroptosis. During PDT-HP treatment, the interaction between CALR and SERCA2 inactivates SERCA2, increasing the susceptibility of cells to increased intracellular Ca2+ levels under oxidative stress. This triggered endoplasmic reticulum stress and activated Caspase3, which further cleaved GSDME, releasing GSDME-N and ultimately leading to pyroptosis in TNBC cells. CONCLUSION In this study, we provide insight into the antitumor mechanism by examining the pharmacological mechanism by which PDT-HP regulates TNBC cell pyroptosis via the ROS/CALR/Caspase-3/GSDME signaling axis.
Collapse
Affiliation(s)
- Chen Liang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Xiao Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Jie Yu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Lingyun Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Wenchao Wei
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Yalu Zhu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Maoping Feng
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Tingting Tang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Dameng Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Tao Yang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China.
| | - Bo Ma
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China.
| | - Liang Wei
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002 Jiangsu, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004 Jiangsu, China.
| |
Collapse
|
4
|
Lü J, Jiang C, Drabick JJ, Joshi M, Perimbeti S. Angelica gigas Nakai (Korean Dang-gui) Root Alcoholic Extracts in Health Promotion and Disease Therapy - active Phytochemicals and In Vivo Molecular Targets. Pharm Res 2025; 42:25-47. [PMID: 39779619 PMCID: PMC11785709 DOI: 10.1007/s11095-024-03809-9] [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: 09/20/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Angelica gigas Nakai (AGN) root is a medicinal herbal widely used in traditional medicine in Korea. AGN root ethanolic extracts have been marketed as dietary supplements in the United States for memory health and pain management. We have recently reviewed the pharmacokinetics (PK) and first-pass hepatic metabolism of ingested AGN supplements in humans for the signature pyranocoumarins decursin (D, Cmax 1x), decursinol angelate (DA, Cmax ~ 10x) and their common botanical precursor and hepatic metabolite decursinol (DOH, Cmax ~ 1000x). Here we update in vivo medicinal activities of AGN and/or its pyranocoumarins and furanocoumarin nodakenin in cancer, pain, memory loss, cerebral ischemia reperfusion stroke, metabolic syndrome and vascular endothelial dysfunctions, anxiety, sleep disorder, epilepsy, inflammatory bowel disease, osteoporosis and osteoarthritis. Given their polypharmacology nature, the pertinent mechanisms of action are likely misrepresented by many cell culture studies that did not consider the drug metabolism knowledge. We report here Rho-associated protein kinases (ROCK1/2) as novel targets for DA and DOH. Combining with published inhibitory activity of DOH on acetylcholinesterase, agonist activity of DOH and antagonist/degrader activity of DA/D on androgen and estrogen receptors, D/DA promoting activity for glutamic acid decarboxylase (GAD)- gamma-aminobutyric acid (GABA) inhibitory axis and inhibition of glutamate dehydrogenase (GDH), monoamine oxidase-A (MAO-A) and transient receptor potential vanilloid 1 (TRPV1), we postulate their contributions to neuro-cognitive, metabolic, oncologic, vascular and other beneficial bioactivities of AGN extracts. A clinical trial is being planned for an AGN extract to manage side effects of androgen deprivation therapy in prostate cancer patients.
Collapse
Affiliation(s)
- Junxuan Lü
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Cannabis and Natural Product Pharmaceutics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Cheng Jiang
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Center for Cannabis and Natural Product Pharmaceutics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Joseph J Drabick
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Monika Joshi
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Stuthi Perimbeti
- Penn State Cancer Institute, Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Medicine Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
5
|
Zong YH, Cao JF, Zhao Y, Gao M, Chen WL, Wu M, Xu X, Xu ZY, Zhang XQ, Tang JZ, Liu Y, Hu XS, Wang SQ, Zhang X. Mechanism of Lian Hua Qing Wen capsules regulates the inflammatory response caused by M 1 macrophage based on cellular experiments and computer simulations. Acta Trop 2024; 257:107320. [PMID: 39002739 DOI: 10.1016/j.actatropica.2024.107320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE The polarization of macrophages with the resulting inflammatory response play a crucial part in tissue and organ damage due to inflammatory. Study has proved Lian Hua Qing Wen capsules (LHQW) can reduce activation of inflammatory response and damage of tissue derived from the inflammatory reactions. However, the mechanism of LHQW regulates the macrophage-induced inflammatory response is unclear. Therefore, we investigated the mechanism of LHQW regulated the inflammatory response of M1 macrophages by cellular experiments and computer simulations. METHODS This study has analysed the targets and mechanisms of macrophage regulating inflammatory response at gene and protein levels through bioinformatics. The monomeric components of LHQW were analyzed by High Performance Liquid Chromatography (HPLC). We established the in vitro cell model by M1 macrophages (Induction of THP-1 cells into M1 macrophages). RT-qPCR and immunofluorescence were used to detect changes in gene and protein levels of key targets after LHQW treatment. Computer simulations were utilized to verify the binding stability of monomeric components and protein targets. RESULTS Macrophages had 140,690 gene targets, inflammatory response had 12,192 gene targets, intersection gene targets were 11,772. Key monomeric components (including: Pinocembrin, Fargesone-A, Nodakenin and Bowdichione) of LHQW were screened by HPLC. The results of cellular experiments indicated that LHQW could significantly reduce the mRNA expression of CCR5, CSF2, IFNG and TNF, thereby alleviating the inflammatory response caused by M1 macrophage. The computer simulations further validated the binding stability and conformation of key monomeric components and key protein targets, and IFNG/Nodakenin was able to form the most stable binding conformation for its action. CONCLUSION In this study, the mechanism of LHQW inhibits the polarization of macrophages and the resulting inflammatory response was investigated by computer simulations and cellular experiments. We found that LHQW may not only reduce cell damage and death by acting on TNF and CCR5, but also inhibit the immune recognition process and inflammatory response by regulating CSF2 and IFNG to prevent polarization of macrophages. Therefore, these results suggested that LHQW may act through multiple targets to inhibit the polarization of macrophages and the resulting inflammatory response.
Collapse
Affiliation(s)
| | - Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu, PR China
| | | | - Miao Gao
- Chengdu Medical College, Chengdu, PR China
| | | | - Mei Wu
- Chengdu Medical College, Chengdu, PR China
| | - Xiang Xu
- Chengdu Medical College, Chengdu, PR China
| | | | | | | | - Yulin Liu
- Chengdu Medical College, Chengdu, PR China
| | | | | | - Xiao Zhang
- Chengdu Medical College, Chengdu, PR China.
| |
Collapse
|
6
|
Zheng X, Tang P, Li H, Ye T, Zhu X, He W, Cheng L, Cheng H. Cucurbitacin E elicits apoptosis in laryngeal squamous cell carcinoma by enhancing reactive oxygen species-regulated mitochondrial dysfunction and endoplasmic reticulum stress. Am J Cancer Res 2024; 14:3905-3921. [PMID: 39267666 PMCID: PMC11387858 DOI: 10.62347/hpqq9412] [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: 06/04/2024] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a prevalent head and neck neoplasm with escalating global morbidity and mortality rates. Despite the increasing burden of LSCC, the drugs currently approved for its treatment are limited. Therefore, it is necessary to identify novel and promising drugs that target LSCC. Cucurbitacin E (CuE) is a naturally oxygenated tetracyclic triterpenoid that suppresses several cancers. However, its anti-LSCC activity and the molecular mechanisms of action remain unclear. This study explored its impact on LSCC, revealing cell viability attenuation and apoptosis enhancement in vitro. Further investigations indicated that CuE significantly decreased mitochondrial membrane potential, thereby promoting cytochrome c release, increasing cleaved-Caspase 3 and cleaved-PARP levels, and triggering mitochondria-dependent apoptosis. Concurrently, exposure of LSCC cells to CuE enhanced endoplasmic reticulum (ER) stress, mobilized the protein kinase RNA-like endoplasmic reticulum kinase/initiation factor 2a/ATF4/C-EBP homologous protein pathway, and induced LSCC cell apoptosis. Finally, CuE markedly elevated intracellular reactive oxygen species (ROS) levels. When ROS were eliminated with N-acetylcysteine, CuE-mediated mitochondrial dysfunction, ER stress, and cell apoptosis were nearly abolished. Similar outcomes were observed in murine LSCC models. Together, these results highlight that CuE suppresses proliferation while triggering apoptosis in LSCC cells via ROS-regulated mitochondrial dysfunction and the ER stress pathway. Hence, CuE may serve as a promising candidate for LCSS treatment.
Collapse
Affiliation(s)
- Xucai Zheng
- Department of Oncology, The Second Hospital of Anhui Medical University Hefei 230601, Anhui, China
- Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital Hefei 230031, Anhui, China
| | - Puze Tang
- Bachelor of Science in Mathematics, University of Liverpool United Kingdom
| | - Hui Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Tingbo Ye
- Department of Head, Neck and Breast Surgery, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital Hefei 230031, Anhui, China
| | - Xu Zhu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Wei He
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University Hefei 230032, Anhui, China
| | - Ling Cheng
- Medical Intensive Care Unit, The First Affiliated Hospital of Anhui University of Chinese Medicine Hefei 230031, Anhui, China
| | - Huaidong Cheng
- Department of Oncology, The Second Hospital of Anhui Medical University Hefei 230601, Anhui, China
- Department of Oncology, Shenzhen Hospital of Southern Medical University Shenzhen 518000, Guangdong, China
| |
Collapse
|
7
|
Hu WB, Liu YT, Li J, Wang Y, Sun XZ, Hua MY, Liu XT, Hui BN. Pristimerin exhibits anti-cancer activity by inducing ER stress and AKT/GSK3β pathway through increasing intracellular ROS production in human esophageal cancer cells. Toxicol In Vitro 2024; 99:105867. [PMID: 38848824 DOI: 10.1016/j.tiv.2024.105867] [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: 02/19/2024] [Revised: 05/13/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Pristimerin (Pris), a bioactive triterpenoid compound extracted from the Celastraceae and Hippocrateaceae families, has been reported to exhibit an anti-cancer property on various cancers. However, the effects of Pris on esophageal cancer are poorly investigated. This current study sought to explore the activity and underlying mechanism of Pris against human esophageal squamous cell carcinoma (ESCC) cells. We demonstrated that Pris showed cytotoxicity in TE-1 and TE-10 ESCC cell lines, and significantly inhibited cell viability in a concentration dependent manner. Pris induced G0/G1 phase arrest and triggered apoptosis. It was also observed that the intracellular ROS level was remarkedly increased by Pris treatment. Besides, the function of Pris mediating the activation of ER stress and the inhibition of AKT/GSK3β signaling pathway in TE-1 and TE-10 cells was further confirmed, which resulted in cell growth inhibition. And moreover, we revealed that all of the above pathways were regulated through ROS generation. In conclusion, our findings suggested that Pris might be considered as a novel natural compound for the developing anti-cancer drug candidate for human esophageal cancer.
Collapse
Affiliation(s)
- Wei-Bin Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yi-Ting Liu
- Department of Medical Oncology, Yan'an University Affiliated Hospital, Yan'an 716000, Shaanxi, China
| | - Jing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ying Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xuan-Zi Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Ming-Yu Hua
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Xue-Ting Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Bei-Na Hui
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
8
|
Chiu FC, Kuo HM, Yu CL, Selvam P, Su IL, Tseng CC, Yuan CH, Wen ZH. Marine-derived antimicrobial peptide piscidin-1 triggers extrinsic and intrinsic apoptosis in oral squamous cell carcinoma through reactive oxygen species production and inhibits angiogenesis. Free Radic Biol Med 2024; 220:28-42. [PMID: 38679300 DOI: 10.1016/j.freeradbiomed.2024.04.235] [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: 03/30/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Cancer of the head and neck encompasses a wide range of cancers, including oral and oropharyngeal cancers. Oral cancer is often diagnosed at advanced stages and has a dismal prognosis. Piscidin-1, a marine antimicrobial peptide (AMP) containing approximately 22 amino acids, also exhibits significant anticancer properties. We investigated the possible anti-oral cancer effects of piscidin-1 and clarified the mechanisms underlying these effects. We treated the oral squamous cell carcinoma cell lines OC2 and SCC4 with piscidin-1. Cell viability and the expression of different hallmark apoptotic molecules, including reactive oxygen species (ROS), were tested using the appropriate MTT assay, flow cytometry and western blotting assays, and human umbilical vein endothelial cell (HUVEC) wound healing, migration, and tube formation (angiogenesis) assays. Piscidin-1 increases cleaved caspase 3 levels to induce apoptosis. Piscidin-1 also increases ROS levels and intensifies oxidative stress in the endoplasmic reticulum and mitochondria, causing mitochondrial dysfunction. Additionally, it decreases the oxygen consumption rates and activity of mitochondrial complexes I-V. As expected, the antioxidants MitoTEMPOL and N-acetylcysteine reduce piscidin-1-induced ROS generation and intracellular calcium accumulation. Piscidin-1 also inhibits matrix metalloproteinase (MMP)-2/-9 expression in HUVECs, affecting migration and tube formation angiogenesis. We demonstrated that piscidin-1 can promote apoptosis via both intrinsic and extrinsic apoptotic pathways and findings indicate that piscidin-1 has anti-proliferative and anti-angiogenic properties in oral cancer treatment. Our study on piscidin-1 thus provides a basis for future translational anti-oral cancer drug research and a new theoretical approach for anti-oral cancer clinical research.
Collapse
Affiliation(s)
- Fu-Ching Chiu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833301, Taiwan
| | - Chen-Ling Yu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Padhmavathi Selvam
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - I-Li Su
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Antai Medical Care Corporation, Antai Tian-Sheng Memorial Hospital, Pingtung, 92842, Taiwan
| | - Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan
| | - Chien-Han Yuan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Department of Otolaryngology, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan; Department of Otolaryngology, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
9
|
NAGINI SIDDAVARAM, KALLAMADI PRATHAPREDDY, TANAGALA KRANTHIKIRANKISHORE, REDDY GEEREDDYBHANUPRAKASH. Aldo-keto reductases: Role in cancer development and theranostics. Oncol Res 2024; 32:1287-1308. [PMID: 39055885 PMCID: PMC11267078 DOI: 10.32604/or.2024.049918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/08/2024] [Indexed: 07/28/2024] Open
Abstract
Aldo-keto reductases (AKRs) are a superfamily of enzymes that play crucial roles in various cellular processes, including the metabolism of xenobiotics, steroids, and carbohydrates. A growing body of evidence has unveiled the involvement of AKRs in the development and progression of various cancers. AKRs are aberrantly expressed in a wide range of malignant tumors. Dysregulated expression of AKRs enables the acquisition of hallmark traits of cancer by activating oncogenic signaling pathways and contributing to chemoresistance. AKRs have emerged as promising oncotherapeutic targets given their pivotal role in cancer development and progression. Inhibition of aldose reductase (AR), either alone or in combination with chemotherapeutic drugs, has evolved as a pragmatic therapeutic option for cancer. Several classes of synthetic aldo-keto reductase (AKR) inhibitors have been developed as potential anticancer agents, some of which have shown promise in clinical trials. Many AKR inhibitors from natural sources also exhibit anticancer effects. Small molecule inhibitors targeting specific AKR isoforms have shown promise in preclinical studies. These inhibitors disrupt the activation of oncogenic signaling by modulating transcription factors and kinases and sensitizing cancer cells to chemotherapy. In this review, we discuss the physiological functions of human AKRs, the aberrant expression of AKRs in malignancies, the involvement of AKRs in the acquisition of cancer hallmarks, and the role of AKRs in oncogenic signaling, and drug resistance. Finally, the potential of aldose reductase inhibitors (ARIs) as anticancer drugs is summarized.
Collapse
|
10
|
Li X, Wang M, Zhong Y, Yin Q, Hu Z, Tian W, Liu Z, Liu Z. Comparative pharmacokinetics of six components in normal and rheumatoid arthritis rats after intragastrical administration of Qianghuo Shengshi Decoction granules by LC-MS/MS. CHINESE HERBAL MEDICINES 2024; 16:457-465. [PMID: 39072204 PMCID: PMC11283214 DOI: 10.1016/j.chmed.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2024] Open
Abstract
Objective To investigate the plasma pharmacokinetics of six representative components (nodakenin, osthole, 5-O-methylvisammioside, ferulic acid, liquiritigenin, and liquiritin), which were the ingredients of Qianghuo Shengshi Decoction (QSD) granules, in normal and rheumatoid arthritis (RA) rats administrated QSD granules intragastrically. Methods A rapid and accurate ultra-high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for the simultaneous determination of six components in plasma, and it showed a good specificity, linearity, intra-day and inter-day precision, intra-day and inter-day accuracy, extraction recovery, stability, and the less matrix effect. Results The validated LC-MS/MS method was successfully used to compare the plasma pharmacokinetics of six ingredients between normal and RA rats after intragastrical administration of QSD granules and differences in the pharmacokinetics were found in two types of rats. The absorption rate in the RA rats was lower for nodakenin, osthole, 5-O-methylvisammioside, liquiritigenin and liquiritin than in the normal group, while the absorption rate of ferulic acid remained constant in two groups. In comparison with the normal rats, the exposure concentration of nodakenin was higher and that of other five components except for nodakenin was lower under pathological conditions. Additionally, the absorptive amount of nodakenin, osthole, 5-O-methylvisammioside and liquiritin was increased and that of ferulic acid and liquiritigenin was reduced in the RA rats than in the normal rats. Compared with the normal rats, the retention time of nodakenin, ferulic acid and liquiritin was reduced in vivo, whereas the retention time of osthole, 5-O-methylvisammioside and liquiritigenin was raised in the body for the RA rats. In contrast to the normal rats, the data demonstrated an increase in the elimination velocity of nodakenin and a decrease in the elimination velocity of the other five components except for nodakenin in the pathological state. Conclusion This study showed that the pharmacokinetic behavior of the six components, nodakenin, osthole, 5-O-methylvisammioside, ferulic acid, liquiritigenin, and liquiritin, is different in vivo between normal and pathological states of rats, and this research provided the necessary experimental data to explain the pharmacokinetics of QSD granules in both normal and pathological states and provide some references for its clinical application at some level.
Collapse
Affiliation(s)
- Xin Li
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Min Wang
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuhong Zhong
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qianqian Yin
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zheming Hu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wenli Tian
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhongyan Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhidong Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
11
|
Kim TW. Paeoniflorin Induces ER Stress-Mediated Apoptotic Cell Death by Generating Nox4-Derived ROS under Radiation in Gastric Cancer. Nutrients 2023; 15:5092. [PMID: 38140352 PMCID: PMC10745742 DOI: 10.3390/nu15245092] [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: 11/08/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Gastric cancer is one of the most prevalent cancer types worldwide, and its resistance to cancer therapies, such as chemotherapy and radiotherapy, has made treating it a major challenge. Paeoniflorin (PF) is one potential pharmacological treatment derived from paeony root. However, in cancer, the molecular mechanisms and biological functions of PF are still unclear. In the present study, we found that PF exerts anti-tumor effects in vivo and in vitro and induces apoptotic cell death through ER stress, calcium (Ca2+), and reactive oxygen species (ROS) release in gastric cancer cells. However, ROS inhibition by DPI and NAC blocks cell death and the PERK signaling pathway via the reduction of Nox4. Moreover, PF triggers a synergistic inhibitory effect of the epithelial-mesenchymal transition (EMT) process under radiation exposure in radiation-resistant gastric cancer cells. These findings indicate that PF-induced Ca2+ and ROS release overcomes radioresistance via ER stress and induces cell death under radiation in gastric cancer cells. Therefore, PF, in combination with radiation, may be a powerful strategy for gastric cancer therapy.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju 38066, Republic of Korea
| |
Collapse
|
12
|
Kim TW. Fisetin, an Anti-Inflammatory Agent, Overcomes Radioresistance by Activating the PERK-ATF4-CHOP Axis in Liver Cancer. Int J Mol Sci 2023; 24:ijms24109076. [PMID: 37240422 DOI: 10.3390/ijms24109076] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/16/2023] [Accepted: 05/20/2023] [Indexed: 05/28/2023] Open
Abstract
Fisetin, a well-known plant flavonol from the natural flavonoid group, is found in traditional medicines, plants, vegetables, and fruits. Fisetin also has anti-oxidant, anti-inflammatory, and anti-tumor effects. This study investigated the anti-inflammatory effects of fisetin in LPS-induced Raw264.7 cells and found that fisetin reduced the LPS-induced production of pro-inflammation markers, such as TNF-α, IL-1β, and IL-6, demonstrating the anti-inflammatory effects of fisetin. Furthermore, this study investigated the anti-cancer effects of fisetin and found that fisetin induced apoptotic cell death and ER stress through intracellular calcium (Ca2+) release, the PERK-ATF4-CHOP signaling pathway, and induction of GRP78 exosomes. However, the suppression of PERK and CHOP inhibited the fisetin-induced cell death and ER stress. Interestingly, fisetin induced apoptotic cell death and ER stress and inhibited the epithelial-mesenchymal transition phenomenon under radiation in radiation-resistant liver cancer cells. These findings indicate that the fisetin-induced ER stress can overcome radioresistance and induce cell death in liver cancer cells following radiation. Therefore, the anti-inflammatory agent fisetin, in combination with radiation, may be a powerful immunotherapy strategy to overcome resistance in an inflammatory tumor microenvironment.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, 123 Dongdae-ro, Gyeongju 38066, Gyeongbuk, Republic of Korea
| |
Collapse
|
13
|
Boonnate P, Kariya R, Okada S. Shikonin Induces ROS-Dependent Apoptosis Via Mitochondria Depolarization and ER Stress in Adult T Cell Leukemia/Lymphoma. Antioxidants (Basel) 2023; 12:864. [PMID: 37107239 PMCID: PMC10135058 DOI: 10.3390/antiox12040864] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/18/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Adult T cell leukemia/lymphoma (ATLL) is an aggressive T-cell malignancy that develops in some elderly human T-cell leukemia virus (HTVL-1) carriers. ATLL has a poor prognosis despite conventional and targeted therapies, and a new safe and efficient therapy is required. Here, we examined the anti-ATLL effect of Shikonin (SHK), a naphthoquinone derivative that has shown several anti-cancer activities. SHK induced apoptosis of ATLL cells accompanied by generation of reactive oxygen species (ROS), loss of mitochondrial membrane potential, and induction of endoplasmic reticulum (ER) stress. Treatment with a ROS scavenger, N-acetylcysteine (NAC), blocked both loss of mitochondrial membrane potential and ER stress, and prevented apoptosis of ATLL cells, indicating that ROS is an upstream trigger of SHK-induced apoptosis of ATLL cells through disruption of the mitochondrial membrane potential and ER stress. In an ATLL xenografted mouse model, SHK treatment suppressed tumor growth without significant adverse effects. These results suggest that SHK could be a potent anti-reagent against ATLL.
Collapse
Affiliation(s)
| | | | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan; (P.B.); (R.K.)
| |
Collapse
|