1
|
Miao Y, Wang X, He F, Zhang F, Huang Y, Lai Y, Wang Y, Zhang L, Yin H, Meng X, Liu H, Li W, Yang S. Targeting CALR reduces energy metabolism of esophageal cancer cells and inhibits tumor‑associated fibroblast infiltration. Int J Oncol 2025; 66:49. [PMID: 40417915 PMCID: PMC12118950 DOI: 10.3892/ijo.2025.5755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/13/2025] [Indexed: 05/27/2025] Open
Abstract
Calreticulin (CALR) supports the induction of dendritic cell maturation, which makes it a key target for effective esophageal squamous cell carcinoma (ESCC) immunotherapy. The mechanism of CALR in the immunotherapy of ESCC is not fully studied. The aim of the present study was to explore the contributing role of CALR in ESCC progression. The association of CALR expression with calnexin (CANX) and protein disulfide isomerase A3 (PDIA3) expression in ESCC was analyzed. The functions of CALR in ESCC cells were examined by detection of cell migration, endoplasmic reticulum (ER) stress, mitochondrial function, cytoskeletal remodeling, cell proliferation and apoptosis. The effects of CALR on tumor growth and tumor‑associated fibroblast infiltration were examined by subcutaneous xenograft assay. The expression of CALR, CANX and PDIA3 in ESCC tissue significantly increased and the expression of PDIA3 was positively associated with CANX. Overexpression of CALR resulted in enhanced cell proliferation, migration, ER stress, mitochondrial function and cytoskeletal remodeling; knockdown of CALR expression had the opposite effect. In the subcutaneous xenograft assay, knockdown CALR significantly inhibited the growth of esophageal cancer tumors, suppressed the invasion of tumor‑associated fibroblasts and decreased the expression of α‑smooth muscle actin (α‑SMA), fibroblast activation protein (FAP), fibroblast specific protein‑1 (FSP1), platelet‑derived growth factor and transforming growth factor beta (TGF‑β) in tumor tissue. These findings suggested that CALR promotes the progression of ESCC by regulating ER stress and mitochondrial function to mediate ATP production, cytoskeletal remodeling, cell proliferation and apoptosis through CANX and PDIA3. Knockdown CALR significantly inhibited tumor‑associated fibroblast infiltration and is a potential drug target for ESCC.
Collapse
Affiliation(s)
- Yu Miao
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Xiaofei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebei 063000, P.R. China
| | - Fang He
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Feixiong Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Ying Huang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Yafang Lai
- Department of Gastroenterology, Ordos Central Hospital, Ordos, Inner Mongolia 017000, P.R. China
| | - Yuanzhen Wang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Lina Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Hua Yin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Xiangkun Meng
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Hao Liu
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Weiqiang Li
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| | - Shaoqi Yang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui 750004, P.R. China
| |
Collapse
|
2
|
Deng Y, Xuan R, Qiu Z, Xiang P, Guo Y, Xu L, Zhang X, Mai H, Li X. Nuclear receptor 4A1 facilitates complete Freund's adjuvant-induced inflammatory pain in rats by promoting ferroptosis in spinal glial cells. Brain Behav Immun 2025; 125:92-109. [PMID: 39722371 DOI: 10.1016/j.bbi.2024.12.152] [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: 07/02/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024] Open
Abstract
Glial cell-induced neuroinflammation in the spinal cord is the critical pathology underlying complete Freund's adjuvant (CFA)-induced inflammatory pain. Previously, we showed that spinal glial cells undergo ferroptosis after CFA injection, which may contribute to the development of neuroinflammation and inflammatory pain. However, the mechanism underlying the occurrence of ferroptosis during inflammatory pain remains unclear. The aim of this study was to investigate the molecular factors involved in the occurrence of ferroptosis during the development of inflammatory pain. Bulk and single-cell RNA sequencing were performed to identify the key genes involved in the ferroptosis of spinal astrocytes, microglia, and oligodendrocytes in rats. We identified nuclear receptor 4A1 (NR4A1) as a common ferroptosis-related gene present in all three types of glial cells. Western blotting and immunostaining revealed increased NR4A1 levels in the spinal glial cells of the CFA-treated rats. Moreover, intrathecal injection of DIM-C-pPhOH (an NR4A1 inhibitor) effectively alleviated mechanical and thermal hypersensitivity in the CFA-treated rats by attenuating ferroptosis and neuroinflammation in spinal glial cells. Proteomic analysis revealed that mitogen-activated protein kinase 3 (MAPK3) may be the target protein of NR4A1. In addition, the combined results of chromatin immunoprecipitation and dual-luciferase assays indicated that NR4A1 can bind to the promoter region and promote transcription of MAPK3, ultimately leading to lipid peroxidation. In conclusion, this study demonstrated that increased expression of NR4A1 promotes the progression of CFA-induced inflammatory pain by enhancing ferroptosis through the transcriptional activation of MAPK3 and subsequent lipid peroxidation. Furthermore, inhibition of NR4A1 was found to suppress ferroptosis and reduce the release of pro-inflammatory cytokines in the spinal cord of rats with inflammatory pain. Collectively, these findings outline a novel pathological mechanism and identify potential therapeutic targets for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Yifan Deng
- Department of Anesthesiology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province 510630, China
| | - Ruoheng Xuan
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510000, China
| | - Zhuolin Qiu
- Department of Anesthesiology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province 510630, China
| | - Ping Xiang
- Department of Medical Quality Management, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yue Guo
- Department of Anesthesiology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province 510630, China
| | - Lejia Xu
- Department of Pharmacy, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province 510630, China
| | - Xiaohan Zhang
- Analysis and Testing Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Haiyan Mai
- Department of Pharmacy, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province 510630, China.
| | - Xiang Li
- Department of Anesthesiology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
3
|
Li H, Gao Y, Ni X, Xiong Y, Zhang P, Liu H, Wu X, Tong D, Wang C, Ma J. Design, synthesis and biological evaluation of 5H-[1,2,4]triazino[5,6-b]indole derivatives bearing a pyridinocycloalkyl moiety as iron chelators. Mol Divers 2025; 29:163-177. [PMID: 38733433 DOI: 10.1007/s11030-024-10840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/07/2024] [Indexed: 05/13/2024]
Abstract
The avidity of cancer cells for iron highlights the potential for iron chelators to be used in cancer therapy. Herein, we designed and synthesized a novel series of 5H-[1,2,4]triazino[5,6-b]indole derivatives bearing a pyridinocycloalkyl moiety using a ring-fusion strategy based on the structure of an iron chelator, VLX600. The antiproliferative activity evaluation against cancer cells and normal cells led to the identification of compound 3k, which displayed the strongest antiproliferative activity in vitro against A549, MCF-7, Hela and HepG-2 with IC50 values of 0.59, 0.86, 1.31 and 0.92 μM, respectively, and had lower cytotoxicity against HEK293 than VLX600. Further investigations revealed that unlike VLX600, compound 3k selectively bound to ferrous ions, but not to ferric ions, and addition of Fe2+ abolished the cytotoxicity of 3k. Flow cytometry assays demonstrated that 3k arrested the cell cycle at the G1 phase and induced significant apoptosis in A549 cells in dose and time-dependent manners, corresponding to JC-1 staining assay results. Western blot analysis of Bcl-2, Bax and cleaved caspase-3 proteins further provided evidences that induction of apoptosis by 3k in A549 cells might be at least via the mitochondria pathway. These above results highlight that 3k is a valuable lead compound that deserves further investigation as an iron chelator for the treatment of cancer.
Collapse
Affiliation(s)
- He Li
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Yali Gao
- Pharmacy Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xin Ni
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Yizu Xiong
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Peixi Zhang
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Han Liu
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Xingye Wu
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Dandan Tong
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China
| | - Cuifang Wang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China.
| | - Junjie Ma
- School of Medicine, Huaqiao University, Quanzhou, 362000, Fujian Province, China.
| |
Collapse
|
4
|
Deng Y, Li Y, Yang M, Gao Y, Luo X, Chen H, Guo M, Yang X, Liu Y, He J, Lu B, Liu N. Carfilzomib activates ER stress and JNK/p38 MAPK signaling to promote apoptosis in hepatocellular carcinoma cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:697-708. [PMID: 38591121 PMCID: PMC11177107 DOI: 10.3724/abbs.2024040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/04/2024] [Indexed: 04/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and deadly cancers in the world, which is frequently diagnosed at a late stage. HCC patients have a poor prognosis due to the lack of an efficacious therapeutic strategy. Approved drug repurposing is a way for accelerating drug discovery and can significantly reduce the cost of drug development. Carfilzomib (CFZ) is a second-generation proteasome inhibitor, which is highly efficacious against multiple myeloma and has been reported to possess potential antitumor activities against multiple cancers. However, the underlying mechanism of CFZ on HCC is still unclear. Here, we show that CFZ inhibits the proliferation of HCC cells through cell cycle arrest at the G2/M phase and suppresses the migration and invasion of HCC cells by inhibiting epithelial-mesenchymal transition. We also find that CFZ promotes reactive oxygen species production to induce endoplasmic reticulum (ER) stress and activate JNK/p38 MAPK signaling in HCC cells, thus inducing cell death in HCC cells. Moreover, CFZ significantly inhibits HCC cell growth in a xenograft mouse model. Collectively, our study elucidates that CFZ impairs mitochondrial function and activates ER stress and JNK/p38 MAPK signaling, thus inhibiting HCC cell and tumor growth. This indicates that CFZ has the potential as a therapeutic drug for HCC.
Collapse
Affiliation(s)
- Yao Deng
- Department of Gastroenterology and Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver DiseaseThe Affiliated Nanhua Hospital and Department of Cell Biology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Yujie Li
- School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhou325035China
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325000China
| | - Mingyue Yang
- School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhou325035China
| | - Yang Gao
- School of Public HealthFudan UniversityShanghai200032China
| | - Xuling Luo
- Department of Gastroenterology and Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver DiseaseThe Affiliated Nanhua Hospital and Department of Cell Biology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Hanbin Chen
- Department of OncologyThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325000China
| | - Meng Guo
- Department of Gastroenterology and Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver DiseaseThe Affiliated Nanhua Hospital and Department of Cell Biology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Xuefeng Yang
- Department of Gastroenterology and Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver DiseaseThe Affiliated Nanhua Hospital and Department of Cell Biology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Yongzhang Liu
- School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhou325035China
| | - Jun He
- Department of Gastroenterology and Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver DiseaseThe Affiliated Nanhua Hospital and Department of Cell Biology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Bin Lu
- Department of Gastroenterology and Hunan Provincial Clinical Research Center for Metabolic Associated Fatty Liver DiseaseThe Affiliated Nanhua Hospital and Department of Cell Biology and GeneticsSchool of Basic Medical SciencesHengyang Medical SchoolUniversity of South ChinaHengyang421001China
- School of Laboratory Medicine and Life SciencesWenzhou Medical UniversityWenzhou325035China
| | - Naxin Liu
- Department of Gastrointestinal SurgeryThe First Affiliated HospitalWenzhou Medical UniversityWenzhou325000China
| |
Collapse
|
5
|
Abdel-Wahab ND, Kabil MF, El-Sherbiny IM, Salama MF, El-Sayed G, El-Sherbini ES. Potential anticancer effect of free and nanoformulated Deferasirox for breast cancer treatment: in-vitro and in-vivo evaluation. Drug Dev Ind Pharm 2024; 50:223-235. [PMID: 38305197 DOI: 10.1080/03639045.2024.2314189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Breast cancer (BC) stands as the second-leading cause of mortality among women worldwide. Many chemotherapeutic treatments for BC come with significant adverse effects. Additionally, BC is recognized as one of the most resistant forms of malignancy to treatment. Consequently, there exists a critical need for innovative therapeutic agents that are both highly effective and exhibit reduced toxicity and side effects for patients. Deferasirox (DFX), an iron-chelating drug approved by the FDA for oral use, emerges as a promising contender in the fight against BC proliferation. DFX, primarily administered orally, is utilized to address chronic iron excess resulting from blood transfusions, and it is the inaugural treatment for chronic iron overload syndrome. However, DFX encounters limitations due to its poor water solubility. AIM This study aimed at incorporating DFX into lipid nanocapsules (DFX-LNCs) followed by investigating the anticancer effect of the DFX nanoform as compared to free DFX in-vitro and on an orthotopic BC mouse model in-vivo. METHODS The DFX-LNCs was prepared and imaged using TEM and also characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI) using DLS. Moreover, drug release, cytotoxicity, and anticancer effect were assessed in-vitro, and in-vivo. RESULTS The results revealed that DFX-LNCs are more cytotoxic than free DFX with IC50 of 4.417 µg/ml and 16.114 µg/ml, respectively, while the plain LNCs didn't show any cytotoxic effect on the 4T1 cell line (IC50 = 122.797 µg/ml). Besides, the apoptotic effect of DFX-LNCs was more pronounced than that of free DFX, as evidenced by Annexin V/PI staining, increased BAX expression, and decreased expression of BcL-2. Moreover, DFX-LNCs showed a superior antitumor effect in-vivo with potent antioxidant and anti-proliferative effects. CONCLUSION The newly developed DFX nanoform demonstrated a high potential as a promising therapeutic agent for BC treatment.
Collapse
Affiliation(s)
- Nadeen Diaa Abdel-Wahab
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Mohamed Fawzi Kabil
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, Giza, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed F Salama
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Gehad El-Sayed
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - El-Said El-Sherbini
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| |
Collapse
|
6
|
Xu L, Peng M, Gao T, Wang D, Lian X, Sun H, Shi J, Wang Y, Wang P. Nanoenabled Intracellular Metal Ion Homeostasis Regulation for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306203. [PMID: 38063781 PMCID: PMC10870045 DOI: 10.1002/advs.202306203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/27/2023] [Indexed: 02/17/2024]
Abstract
Endogenous essential metal ions play an important role in many life processes, especially in tumor development and immune response. The approval of various metallodrugs for tumor therapy brings more attention to the antitumor effect of metal ions. With the deepening understanding of the regulation mechanisms of metal ion homeostasis in vivo, breaking intracellular metal ion homeostasis becomes a new means to inhibit the proliferation of tumor cells and activate antitumor immune response. Diverse nanomedicines with the loading of small molecular ion regulators or metal ions have been developed to disrupt metal ion homeostasis in tumor cells, with higher safety and efficiency than free small molecular ion regulators or metal compounds. This comprehensive review focuses on the latest progress of various intracellular metal ion homeostasis regulation-based nanomedicines in tumor therapy including calcium ion (Ca2+ ), ferrous ion (Fe2+ ), cuprous ion (Cu+ ), managanese ion (Mn2+ ), and zinc ion (Zn2+ ). The physiological functions and homeostasis regulation processes of ions are summarized to guide the design of metal ion regulation-based nanomedicines. Then the antitumor mechanisms of various ions-based nanomedicines and some efficient synergistic therapies are highlighted. Finally, the challenges and future developments of ion regulation-based antitumor therapy are also discussed, hoping to provide a reference for finding more effective metal ions and synergistic therapies.
Collapse
Affiliation(s)
- Lihua Xu
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Mingzheng Peng
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Tingting Gao
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
| | - Dandan Wang
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Xiaowu Lian
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhou450052China
| | - Huihui Sun
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Jinjin Shi
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
| | - Yafeng Wang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
| | - Pengju Wang
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| |
Collapse
|
7
|
Wang HG, Wang D, Sarfraz M, Afzal A, Jing MR, Zhang YX, Cai CB, Qi HW, Chen HJ, Li T, Hu SJ, Liu HX, Ji XY, Wu DD. Endogenous hydrogen sulfide inhibition suppresses tumor growth by promoting apoptosis and pyroptosis in esophageal cancer cells. Transl Oncol 2023; 38:101770. [PMID: 37716259 PMCID: PMC10514559 DOI: 10.1016/j.tranon.2023.101770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 08/10/2023] [Accepted: 08/21/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Hydrogen sulfide (H2S) has been identified as the third gaseous signaling molecule. Endogenous H2S plays a key role in the progression of various types of cancer. However, the effect of endogenous H2S on the growth of esophageal cancer (EC) remains unknown. METHODS In this study, three kinds of H2S-producing enzymes inhibitors, DL-propargylglycine (PAG, inhibitor of cystathionine-γ-lyase), aminooxyacetic acid (AOAA, inhibitor of cystathionine-β-synthase), and L-aspartic acid (L-Asp, inhibitor of 3-mercaptopyruvate sulfurtransferase) were used to determine the role of endogenous H2S in the growth of EC9706 and K450 human EC cells. RESULTS The results indicated that the combination (PAG+AOAA+L-Asp) group showed higher inhibitory effects on the viability, proliferation, migration, and invasion of EC cells than PAG, AOAA, and L-Asp group. Inhibition of endogenous H2S promoted apoptosis via activation of mitogen-activated protein kinase pathway in EC cells. Endogenous H2S suppression triggered pyroptosis of EC cells by activating reactive oxygen species-mediated nuclear factor-κB signaling pathway. In addition, the combine group showed its more powerful growth-inhibitory effect on the growth of human EC xenograft tumors in nude mice without obvious toxicity. CONCLUSION Our results indicate that inhibition of endogenous H2S production can significantly inhibit human EC cell growth via promotion of apoptosis and pyroptosis. Endogenous H2S may be a promising therapeutic target in EC cells. Novel inhibitors for H2S-producing enzymes can be designed and developed for EC treatment.
Collapse
Affiliation(s)
- Hong-Gang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Di Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Muhammad Sarfraz
- Department of Science, South East Technological University, Main Campus Waterford X91K0EK, Ireland; School of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Lahore, Lahore 56400, Pakistan
| | - Attia Afzal
- School of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Lahore, Lahore 56400, Pakistan
| | - Mi-Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Chun-Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Hui-Wen Qi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Hao-Jie Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Tao Li
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan 475004, China; Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shui-Juan Hu
- School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Hong-Xia Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, Henan 475004, China; Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China.
| |
Collapse
|
8
|
Xi Y, Shen Y, Chen L, Tan L, Shen W, Niu X. Exosome-mediated metabolic reprogramming: Implications in esophageal carcinoma progression and tumor microenvironment remodeling. Cytokine Growth Factor Rev 2023; 73:78-92. [PMID: 37696716 DOI: 10.1016/j.cytogfr.2023.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023]
Abstract
Esophageal carcinoma is among the most fatal malignancies with increasing incidence globally. Tumor onset and progression can be driven by metabolic reprogramming, especially during esophageal carcinoma development. Exosomes, a subset of extracellular vesicles, display an average size of ∼100 nanometers, containing multifarious components (nucleic acids, proteins, lipids, etc.). An increasing number of studies have shown that exosomes are capable of transferring molecules with biological functions into recipient cells, which play crucial roles in esophageal carcinoma progression and tumor microenvironment that is a highly heterogeneous ecosystem through rewriting the metabolic processes in tumor cells and environmental stromal cells. The review introduces the reprogramming of glucose, lipid, amino acid, mitochondrial metabolism in esophageal carcinoma, and summarize current pharmaceutical agents targeting such aberrant metabolism rewiring. We also comprehensively overview the biogenesis and release of exosomes, and recent advances of exosomal cargoes and functions in esophageal carcinoma and their promising clinical application. Moreover, we discuss how exosomes trigger tumor growth, metastasis, drug resistance, and immunosuppression as well as tumor microenvironment remodeling through focusing on their capacity to transfer materials between cells or between cells and tissues and modulate metabolic reprogramming, thus providing a theoretical reference for the design potential pharmaceutical agents targeting these mechanisms. Altogether, our review attempts to fully understand the significance of exosome-based metabolic rewriting in esophageal carcinoma progression and remodeling of the tumor microenvironment, bringing novel insights into the prevention and treatment of esophageal carcinoma in the future.
Collapse
Affiliation(s)
- Yong Xi
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China; Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yaxing Shen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lijie Chen
- School of Medicine, Xiamen University, Xiamen 361102, Fujian, China; China Medical University, Shenyang 110122, Liaoning, China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Weiyu Shen
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China.
| | - Xing Niu
- China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
9
|
Varlı M, Kim SJ, Noh MG, Kim YG, Ha HH, Kim KK, Kim H. KITENIN promotes aerobic glycolysis through PKM2 induction by upregulating the c-Myc/hnRNPs axis in colorectal cancer. Cell Biosci 2023; 13:146. [PMID: 37553596 PMCID: PMC10410973 DOI: 10.1186/s13578-023-01089-1] [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: 03/02/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023] Open
Abstract
PURPOSE The oncoprotein KAI1 C-terminal interacting tetraspanin (KITENIN; vang-like 1) promotes cell metastasis, invasion, and angiogenesis, resulting in shorter survival times in cancer patients. Here, we aimed to determine the effects of KITENIN on the energy metabolism of human colorectal cancer cells. EXPERIMENTAL DESIGN The effects of KITENIN on energy metabolism were evaluated using in vitro assays. The GEPIA web tool was used to extrapolate the clinical relevance of KITENIN in cancer cell metabolism. The bioavailability and effect of the disintegrator of KITENIN complex compounds were evaluated by LC-MS, in vivo animal assay. RESULTS KITENIN markedly upregulated the glycolytic proton efflux rate and aerobic glycolysis by increasing the expression of GLUT1, HK2, PKM2, and LDHA. β-catenin, CD44, CyclinD1 and HIF-1A, including c-Myc, were upregulated by KITENIN expression. In addition, KITENIN promoted nuclear PKM2 and PKM2-induced transactivation, which in turn, increased the expression of downstream mediators. This was found to be mediated through an effect of c-Myc on the transcription of hnRNP isoforms and a switch to the M2 isoform of pyruvate kinase, which increased aerobic glycolysis. The disintegration of KITENIN complex by silencing the KITENIN or MYO1D downregulated aerobic glycolysis. The disintegrator of KITENIN complex compound DKC1125 and its optimized form, DKC-C14S, exhibited the inhibition activity of KITENIN-mediated aerobic glycolysis in vitro and in vivo. CONCLUSIONS The oncoprotein KITENIN induces PKM2-mediated aerobic glycolysis by upregulating the c-Myc/hnRNPs axis.
Collapse
Affiliation(s)
- Mücahit Varlı
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam, 57922, Republic of Korea
| | - Sung Jin Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam, 57922, Republic of Korea
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwangju, 61469, Republic of Korea
| | - Myung-Giun Noh
- Department of Pathology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwanju, 61469, Republic of Korea
| | - Yoon Gyoon Kim
- College of Pharmacy, Dankook University, 119 Dandaero, Dongnam-gu, 31116, Cheonan-si, Republic of Korea
| | - Hyung-Ho Ha
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam, 57922, Republic of Korea
| | - Kyung Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseoro, Dong-gu, Gwangju, 61469, Republic of Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam, 57922, Republic of Korea.
| |
Collapse
|
10
|
Zhang H, Shangguan F, Zhang L, Ma N, Song S, Ma L, Liu C, Liu M, An J, Li H, Cao Q. A novel mechanism of 6-methoxydihydroavicine in suppressing ovarian carcinoma by disrupting mitochondrial homeostasis and triggering ROS/ MAPK mediated apoptosis. Front Pharmacol 2023; 14:1093650. [PMID: 37214469 PMCID: PMC10196025 DOI: 10.3389/fphar.2023.1093650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction: Alkaloids derived from M. cordata (Papaveraceae family), have been found to display antineoplastic activity in several types of cancer. However, the antitumor effects and mechanisms of a new alkaloid extracted from the fruits of M. cordata, named 6-Methoxydihydroavicine (6-ME), remains unclear in the case of ovarian cancer (OC). Methods: CCK-8 assay was employed to analyze the cell viabilities of OC cells. RTCA, and colony-formation assays were performed to measure OC cell growth. Alterations in apoptosis and ROS levels were detected by flow cytometry in accordance with the instructions of corresponding assay kits. A Seahorse XFe96 was executed conducted to confirm the effects of 6-ME on cellular bioenergetics. Western blot and q-RT-PCR were conducted to detect alterations in target proteins. The subcutaneous xenografted tumor model of OC was used to further validate the anti-tumor activity of 6-ME in vivo. Results: Here, we reported for the first time that 6-ME inhibits OC cells growth in vitro and in vivo. Meanwhile, we found that 6-ME showed great antineoplastic activities by disrupting mitochondria homeostasis and promoting apoptosis in OC cells. Further investigation of the upstream signaling of apoptosis revealed that 6-ME-triggered apoptosis was induced by reactive oxygen species (ROS)-mediated mitogen-activated protein kinase (MAPK) activation and mitochondria dysfunction in OC cells. Furthermore, we found oxaloacetic acid (OAA), a crucial metabolite has been proved to be related to NADPH production, can block the cytotoxicity and accumulation of ROS caused by 6-ME in OC cells. Discussion: In summary, our data show that 6-ME exhibits cytotoxicity to OC cells in a ROS-dependent manner by interrupting mitochondrial respiration homeostasis and inducing MAPK-mediated apoptosis. This evidence suggests that 6-ME is a promising remedy for OC intervention.
Collapse
Affiliation(s)
- Huachang Zhang
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China
| | - Fugen Shangguan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lan Zhang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Nengfang Ma
- School of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Shuling Song
- School of Gerontology, Binzhou Medical University, Yantai, Shandong, China
| | - Li Ma
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China
| | - Chuntong Liu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China
| | - Mengke Liu
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China
| | - Jing An
- Division of Infectious Diseases and Global Health, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, United States
| | - Hua Li
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Qizhi Cao
- Department of Immunology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
11
|
Zhou J, Yang Y, Cheng J, Luan S, Xiao X, Li X, Fang P, Gu Y, Shang Q, Zhang H, Chen L, Zeng X, Yuan Y. MTHFD1L confers a poor prognosis and malignant phenotype in esophageal squamous cell carcinoma by activating the ERK5 signaling pathway. Exp Cell Res 2023; 427:113584. [PMID: 37004948 DOI: 10.1016/j.yexcr.2023.113584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/05/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
MTHFD1L, a key enzyme of folate metabolism, is seldom reported in cancer. In this study, we investigate the role of MTHFD1L in the tumorigenicity of esophageal squamous cell carcinoma (ESCC). ESCC tissue microarrays (TMAs) containing samples from 177 patients were utilized to evaluate whether MTHFD1L expression, determined using immunohistochemical analysis, is a prognostic indicator for ESCC patients. The function of MTHFD1L in the migration of ESCC cells was studied with wound healing, Transwell, and three-dimensional spheroid invasion assays in vitro and a lung metastasis mouse model in vivo. The mRNA microarrays and Ingenuity pathway analysis (IPA) were used to explore the downstream of MTHFD1L. Elevated expression of MTHFD1L in ESCC tissues was significantly associated with poor differentiation and prognosis. These phenotypic assays revealed that MTHFD1L significantly promote the viability and metastasis of ESCC cell in vivo and in vitro. Further detailed analyses of the molecular mechanism demonstrated that the ESCC progression driven by MTHFD1L was through up-regulation ERK5 signaling pathways. These findings reveal that MTHFD1L is positively associated with the aggressive phenotype of ESCC by activating ERK5 signaling pathways, suggesting that MTHFD1L is a new biomarker and a potential molecular therapeutic target for ESCC.
Collapse
Affiliation(s)
- Jianfeng Zhou
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yushang Yang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahan Cheng
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Siyuan Luan
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Xiao
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaokun Li
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Pinhao Fang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yimin Gu
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qixin Shang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hanlu Zhang
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Longqi Chen
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoxi Zeng
- Biomedical Big Data Center of West China Hospital, Med+X Center for Informatics, Sichuan University, Chengdu, 610041, China
| | - Yong Yuan
- Department of Thoracic Surgery, Med+X Center for Informatics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Lu J, Li Y, Gong S, Wang J, Lu X, Jin Q, Lu B, Chen Q. Ciclopirox targets cellular bioenergetics and activates ER stress to induce apoptosis in non-small cell lung cancer cells. Cell Commun Signal 2022; 20:37. [PMID: 35331268 PMCID: PMC8943949 DOI: 10.1186/s12964-022-00847-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 02/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background Lung cancer remains a major cause of cancer-related mortality throughout the world at present. Repositioning of existing drugs for other diseases is a promising strategy for cancer therapies, which may rapidly advance potentially promising agents into clinical trials and cut down the cost of drug development. Ciclopirox (CPX), an iron chelator commonly used to treat fungal infections, which has recently been shown to have antitumor activity against a variety of cancers including both solid tumors and hematological malignancies in vitro and in vivo. However, the effect of CPX on non-small cell lung cancer (NSCLC) and the underlying mechanism is still unclear. Methods CCK-8, clonal formation test and cell cycle detection were used to observe the effect of inhibitor on the proliferation ability of NSCLC cells. The effects of CPX on the metastasis ability of NSCLC cells were analyzed by Transwell assays. Apoptosis assay was used to observe the level of cells apoptosis. The role of CPX in energy metabolism of NSCLC cells was investigated by reactive oxygen species (ROS) detection, glucose uptake, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) experiments. Western blot was used to examine the protein changes. Results We report that CPX inhibits NSCLC cell migration and invasion abilities through inhibiting the epithelial-mesenchymal transition, impairing cellular bioenergetics, and promoting reactive oxygen species to activate endoplasmic reticulum (ER) stress-induced apoptotic cell death. Moreover, CPX intraperitoneal injection can significantly inhibit NSCLC growth in vivo in a xenograft model. Conclusions Our study revealed that CPX targets cellular bioenergetics and activates unfolded protein response in ER to drive apoptosis in NSCLC cells, indicating that CPX may be a potential therapeutic drug for the treatment of NSCLC. Graphical Abstract ![]()
Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00847-x.
Collapse
Affiliation(s)
- Junwan Lu
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,School of Medicine, Jinhua Polytechnic, Jinhua, 321007, China
| | - Yujie Li
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shiwei Gong
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,Department of Laboratory Medicine, Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430030, Hubei, China
| | - Jiaxin Wang
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiaoang Lu
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Qiumei Jin
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Bin Lu
- Protein Quality Control and Diseases Laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Qin Chen
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
13
|
Pang Y, Lin Y, Wang X, Wang J, Liu Q, Ding N, Huang L, Xiang Q, Fang J, Tan G, Lyu J, Wang Z. Inhibition of abnormally activated HIF-1α-GLUT1/3-glycolysis pathway enhances the sensitivity of hepatocellular carcinoma to 5-caffeoylquinic acid and its derivatives. Eur J Pharmacol 2022; 920:174844. [DOI: 10.1016/j.ejphar.2022.174844] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 11/03/2022]
|
14
|
Jin G, Ruan Q, Shangguan F, Lan L. RUNX2 and LAMC2: promising pancreatic cancer biomarkers identified by an integrative data mining of pancreatic adenocarcinoma tissues. Aging (Albany NY) 2021; 13:22963-22984. [PMID: 34606473 PMCID: PMC8544338 DOI: 10.18632/aging.203589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/18/2021] [Indexed: 01/25/2023]
Abstract
Pancreatic carcinoma (PC) is a severe disease associated with high mortality. Although strategies for cancer therapy have made great progress, outcomes of pancreatic carcinoma patients remain extremely poor. Therefore, it is urgent to find novel biomarkers and therapeutic targets. To identify biomarkers for early diagnosis and therapy, three mRNA microarray datasets and two miRNA datasets were selected, and combinative analysis was performed by GEO2R. Functional and pathway enrichment analysis were performed using DAVID database. MiRTarBase, miRWalk and Diana Tools were used to get key genes. TCGA, HPA and western blotting were used to verify diagnostic and prognostic value of key genes. By integrating mRNA and miRNA expression profiles, we identified 114 differentially expressed genes and 114 differentially expressed miRNAs, respectively. Then, three overlapping key genes, RUNX2, LAMC2 and FBXO32, were found. Their protein levels in pancreatic tissue from PC patients and normal people were analyzed by immunohistochemical staining and western blotting. RUNX2 showed a potential property to identify PC. Aberrant over-expression of LAMC2 was associated with poor prognosis of PC patients, tumor status and subtypes. In summary, our current study identified that RUNX2 and LAMC2 may be promising targets for early diagnosis and therapy of PC patients.
Collapse
Affiliation(s)
- Guihua Jin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qingqing Ruan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Fugen Shangguan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
15
|
Taş İ, Varlı M, Son Y, Han J, Kwak D, Yang Y, Zhou R, Gamage CDB, Pulat S, Park SY, Yu YH, Moon KS, Lee KH, Ha HH, Hur JS, Kim H. Physciosporin suppresses mitochondrial respiration, aerobic glycolysis, and tumorigenesis in breast cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153674. [PMID: 34333327 DOI: 10.1016/j.phymed.2021.153674] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 05/25/2023]
Abstract
BACKGROUND Physciosporin (PHY) is one of the potent anticancer lichen compound. Recently, PHY was shown to suppress colorectal cancer cell proliferation, motility, and tumorigenesis through novel mechanisms of action. PURPOSE We investigated the effects of PHY on energy metabolism and tumorigenicity of the human breast cancer (BC) cells MCF-7 (estrogen and progesterone positive BC) and MDA-MB-231 (triple negative BC). METHODS The anticancer effect of PHY on cell viability, motility, cancer metabolism and tumorigenicity was evaluated by MTT assay, migration assay, clonogenic assay, anchorage-independent colony formation assay, glycolytic and mitochondrial metabolism analysis, qRT-PCR, flow cytometric analysis, Western blotting, immunohistochemistry in vitro; and by tumorigenicity study with orthotopic breast cancer xenograft model in vivo. RESULTS PHY markedly inhibited BC cell viability. Cell-cycle profiling and Annexin V-FITC/PI double staining showed that a toxic dosage of PHY triggered apoptosis in BC cell lines by regulating the B-cell lymphoma-2 (Bcl-2) family proteins and the activity of caspase pathway. At non-toxic concentrations, PHY potently decreased migration, proliferation, and tumorigenesis of BC cells in vitro. Metabolic studies revealed that PHY treatment significantly reduced the bioenergetic profile by decreasing respiration, ATP production, and glycolysis capacity. In addition, PHY significantly altered the levels of mitochondrial (PGC-1α) and glycolysis (GLUT1, HK2 and PKM2) markers, and downregulated transcriptional regulators involved in cancer cell metabolism, including β-catenin, c-Myc, HIF-1α, and NF-κB. An orthotopic implantation mouse model of BC confirmed that PHY treatment suppressed BC growth in vivo and target genes were consistently suppressed in tumor specimens. CONCLUSION The findings from our in vitro as well as in vivo studies exhibit that PHY suppresses energy metabolism as well as tumorigenesis in BC. Especially, PHY represents a promising therapeutic effect against hormone-insensitive BC (triple negative) by targeting energy metabolism.
Collapse
Affiliation(s)
- İsa Taş
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea; Korean Lichen Research Institute, Sunchon National University, Sunchon, Republic of Korea
| | - Mücahit Varlı
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Yeseon Son
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Jin Han
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Dahye Kwak
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Yi Yang
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | | | - Sultan Pulat
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - So-Yeon Park
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Young Hyun Yu
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Kyung-Sub Moon
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyung-Ho Ha
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea
| | - Jae-Seoun Hur
- Korean Lichen Research Institute, Sunchon National University, Sunchon, Republic of Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon, Republic of Korea.
| |
Collapse
|
16
|
Hui Y, Tang T, Wang J, Zhao H, Yang HY, Xi J, Zhang B, Fang J, Gao K, Wu Y. Fusaricide is a Novel Iron Chelator that Induces Apoptosis through Activating Caspase-3. JOURNAL OF NATURAL PRODUCTS 2021; 84:2094-2103. [PMID: 34292737 DOI: 10.1021/acs.jnatprod.0c01322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nonsmall cell lung cancer (NSCLC) has been a fatal and refractory disease worldwide. Novel therapeutic developments based on fundamental investigations of anticancer mechanisms underlie substantial foundations to win the fight against cancer diseases. In this study, we isolated a natural product fusaricide (FCD) from an endophytic fungus of Lycium barbarum, identified as Epicoccum sp. For the first time, we discovered that FCD potently inhibited proliferation in a variety of human NSCLC cell lines, with relatively less toxicity to normal cells. Our study exhibited that FCD induced apoptosis, caused DNA damage and cell cycle arrest in G0/G1 phase, and activated caspase-3 as well as other apoptosis-related factors in human NSCLC NCI-H460 cells. FCD was proven to be an iron chelator that actively decreased levels of cellular labile iron pool in NCI-H460 cells in our study. FeCl3 supplement reversed FCD-induced apoptosis. The upregulation of transferrin receptor 1 (TfR1) and downregulation of ferritin heavy chain (FTH) expression were observed after FCD treatment. In summary, our study highlighted the potential anticancer effects of FCD against human NSCLCs and demonstrated that the FCD-mediated apoptosis depended on binding to intracellular iron.
Collapse
Affiliation(s)
- Yaling Hui
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Ting Tang
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Jing Wang
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Huanhuan Zhao
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Hong-Ying Yang
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Junmin Xi
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Kun Gao
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Yueting Wu
- State Key Laboratory of Applied Organic Chemistry (SKLAOC), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| |
Collapse
|
17
|
Cisplatin-induced hydroxyl radicals mediate pro-survival autophagy in human lung cancer H460 cells. Biol Res 2021; 54:22. [PMID: 34321115 PMCID: PMC8317380 DOI: 10.1186/s40659-021-00346-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Background Accumulated evidence demonstrates cisplatin, a recommended chemotherapy, modulating pro-survival autophagic response that contributes to treatment failure in lung cancer patients. However, distinct mechanisms involved in cisplatin-induced autophagy in human lung cancer cells are still unclear. Results Herein, role of autophagy in cisplatin resistance was indicated by a decreased cell viability and increased apoptosis in lung cancer H460 cells pre-incubated with wortmannin, an autophagy inhibitor, prior to treatment with 50 µM cisplatin for 24 h. The elevated level of hydroxyl radicals detected via flow-cytometry corresponded to autophagic response, as evidenced by the formation of autophagosomes and autolysosomes in cisplatin-treated cells. Interestingly, apoptosis resistance, autophagosome formation, and the alteration of the autophagic markers, LC3-II/LC3-I and p62, as well as autophagy-regulating proteins Atg7 and Atg3, induced by cisplatin was abrogated by pretreatment of H460 cells with deferoxamine, a specific hydroxyl radical scavenger. The modulations in autophagic response were also indicated in the cells treated with hydroxyl radicals generated via Fenton reaction, and likewise inhibited by pretreatment with deferoxamine. Conclusions In summary, the possible role of hydroxyl radicals as a key mediator in the autophagic response to cisplatin treatment, which was firstly revealed in this study would benefit for the further development of novel therapies for lung cancer.
Collapse
|
18
|
Zhu Y, Chang J, Tan K, Huang SK, Liu X, Wang X, Cao M, Zhang H, Li S, Duan X, Chang Y, Fan Y, Cao P. Clioquinol Attenuates Pulmonary Fibrosis through Inactivation of Fibroblasts via Iron Chelation. Am J Respir Cell Mol Biol 2021; 65:189-200. [PMID: 33861690 DOI: 10.1165/rcmb.2020-0279oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Strict control of iron homeostasis is critical for the maintenance of normal lung function. Iron accumulates in the lungs of patients with idiopathic pulmonary fibrosis (PF), but the characteristics of iron metabolism in the pathogenesis of PF and related targeting therapeutics are not well studied. In this study, we investigated the cellular and molecular characteristics of iron metabolism in fibrotic lungs and further explored the efficacy of clioquinol (CQ) for the treatment of PF as well as its functional mechanism. Iron aggregates accumulated in the lungs of patients with idiopathic PF, and FTL (ferritin light chain) transcripts were increased in their pulmonary fibroblasts. In the bleomycin (BLM)-induced PF (BLM-PF) mouse model, pulmonary iron accumulation is a very early and concomitant event of PF. Labile iron pool levels in both fibroblasts and macrophages from the BLM-PF model were elevated, and iron metabolism was dysregulated. CQ attenuated PF induced by BLM and FITC, and iron-saturated CQ did not alleviate BLM-PF. Furthermore, CQ inhibited the activation of fibroblasts, including proliferation, fibrotic differentiation, proinflammatory cytokine secretion, and migration. In conclusion, our study demonstrated that CQ, acting as an iron chelator, attenuates experimental PF through inactivation of fibroblasts, providing support for targeting iron metabolism as a basis for PF treatment.
Collapse
Affiliation(s)
- Yumeng Zhu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Jing Chang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Ke Tan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, University of Michigan, Ann Arbor, Michigan; and
| | - Xin Liu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Xiaofan Wang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Mengshu Cao
- Department of Respiratory and Critical Care Medicine, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Hongmin Zhang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Shuxin Li
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Xianglin Duan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yanzhong Chang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Yumei Fan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Pengxiu Cao
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Su Z, Han S, Jin Q, Zhou N, Lu J, Shangguan F, Yu S, Liu Y, Wang L, Lu J, Li Q, Cai L, Wang C, Tian X, Chen L, Zheng W, Lu B. Ciclopirox and bortezomib synergistically inhibits glioblastoma multiforme growth via simultaneously enhancing JNK/p38 MAPK and NF-κB signaling. Cell Death Dis 2021; 12:251. [PMID: 33674562 PMCID: PMC7935936 DOI: 10.1038/s41419-021-03535-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 02/05/2023]
Abstract
Ciclopirox (CPX) is an antifungal drug that has recently been reported to act as a potential anticancer drug. However, the effects and underlying molecular mechanisms of CPX on glioblastoma multiforme (GBM) remain unknown. Bortezomib (BTZ) is the first proteasome inhibitor-based anticancer drug approved to treat multiple myeloma and mantle cell lymphoma, as BTZ exhibits toxic effects on diverse tumor cells. Herein, we show that CPX displays strong anti-tumorigenic activity on GBM. Mechanistically, CPX inhibits GBM cellular migration and invasion by reducing N-Cadherin, MMP9 and Snail expression. Further analysis revealed that CPX suppresses the expression of several key subunits of mitochondrial enzyme complex, thus leading to the disruption of mitochondrial oxidative phosphorylation (OXPHOS) in GBM cells. In combination with BTZ, CPX promotes apoptosis in GBM cells through the induction of reactive oxygen species (ROS)-mediated c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK) signaling. Moreover, CPX and BTZ synergistically activates nuclear factor kappa B (NF-κB) signaling and induces cellular senescence. Our findings suggest that a combination of CPX and BTZ may serve as a novel therapeutic strategy to enhance the anticancer activity of CPX against GBM.
Collapse
Affiliation(s)
- Zhipeng Su
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shengnan Han
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Pathology, The Second Hospital of Jiaxing, Jiaxing University, Jiaxing, 314000, China
| | - Qiumei Jin
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ningning Zhou
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Junwan Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fugen Shangguan
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shiyi Yu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yongzhang Liu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lu Wang
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jianglong Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Li
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lin Cai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chengde Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaohe Tian
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and molecular imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Lingyan Chen
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Weiming Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Bin Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
20
|
Abstract
Cancer cells accumulate iron to supplement their aberrant growth and metabolism. Depleting cells of iron by iron chelators has been shown to be selectively cytotoxic to cancer cells in vitro and in vivo. Iron chelators are effective at combating a range of cancers including those which are difficult to treat such as androgen insensitive prostate cancer and cancer stem cells. This review will evaluate the impact of iron chelation on cancer cell survival and the underlying mechanisms of action. A plethora of studies have shown iron chelators can reverse some of the major hallmarks and enabling characteristics of cancer. Iron chelators inhibit signalling pathways that drive proliferation, migration and metastasis as well as return tumour suppressive signalling. In addition to this, iron chelators stimulate apoptotic and ER stress signalling pathways inducing cell death even in cells lacking a functional p53 gene. Iron chelators can sensitise cancer cells to PARP inhibitors through mimicking BRCAness; a feature of cancers trademark genomic instability. Iron chelators target cancer cell metabolism, attenuating oxidative phosphorylation and glycolysis. Moreover, iron chelators may reverse the major characteristics of oncogenic transformation. Iron chelation therefore represent a promising selective mode of cancer therapy.
Collapse
|
21
|
Yang H, Said AM, Huang H, Papa APD, Jin G, Wu S, Ma N, Lan L, Shangguan F, Zhang Q. Chlorogenic acid depresses cellular bioenergetics to suppress pancreatic carcinoma through modulating c-Myc-TFR1 axis. Phytother Res 2020; 35:2200-2210. [PMID: 33258205 DOI: 10.1002/ptr.6971] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 10/26/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is severe malignant tumor in human, the outcomes of PDAC is extremely poor. Here, we evaluated the potential anti-tumor activity of chlorogenic Acid (CA) in PDAC. Here, we found CA was effective to suppress PDAC cell growth in vitro and in vivo. Importantly, we found overall oxygen consumption rate was significantly decreased in CA dose-dependent manner. We also found glycolysis reverse was decreased in CA-treated cells, while basal glycolysis and glycolytic capacity were not significantly changed. Mechanistically, we demonstrated TFR1 could be a novel downstream target of CA, which is essential for PDAC cell growth and cellular bioenergetics maintenance. Furthermore, we validated that CA-reduced c-Myc resulted to down-regulation of TFR1, which contributes to mitochondrial respiration dysfunction and cell growth delay. Together, this study indicates that CA suppresses PDAC cell growth through targeting c-Myc-TFR1 axis and suggests CA could be considered as a promising compound for PDAC treatment.
Collapse
Affiliation(s)
- Hongbao Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Abdullahi Mohamed Said
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huimin Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Akuetteh Percy David Papa
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guihua Jin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shijia Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Nengfang Ma
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fugen Shangguan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiyu Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
22
|
Zhao Y, Li J, Guo W, Li H, Lei L. Periodontitis-level butyrate-induced ferroptosis in periodontal ligament fibroblasts by activation of ferritinophagy. Cell Death Discov 2020; 6:119. [PMID: 33298848 PMCID: PMC7655826 DOI: 10.1038/s41420-020-00356-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/08/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Loss of periodontal ligament fibroblasts (PDLFs) is one critical issue for regenerating lost periodontal tissues. A wide variety of regulated cell death pathways, such as apoptosis, pyroptosis, and necroptosis have been proposed in the periodontitis development. The aim of the present study was to explore whether long-term periodontitis-level butyrate may trigger ferroptosis, a newly characterized iron-dependent regulated cell death in PDLFs. Here, we showed that long-term treatment of butyrate, an important short-chain fatty acid in the periodontal pocket, induces the cargo receptor nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and ferroptosis in PDLFs. Butyrate-induced iron accumulation, reactive oxygen species (ROS) generation, glutathione depletion and lipid peroxidation in PDLFs, and the butyrate-induced ferroptosis can be blocked by the lipid peroxide scavenger ferrostatin-1. The NCOA4-mediated ferritinophagy is dependent on p38/hypoxia inducible factor-1α (HIF-1α) pathway activation as well as Bromodomain-containing protein (BRD) 4 and cyclin-dependent kinase 9 (CDK9) coordination. These lines of evidence provide a new mechanistic insight into the mechanism of loss of PDLFs during periodontitis development, showing that periodontitis-level butyrate disrupted iron homeostasis by activation of NCOA4-mediated ferritinophagy, leading to ferroptosis in PDLFs.
Collapse
Affiliation(s)
- Yunhe Zhao
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Jiao Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Wei Guo
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Houxuan Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.
| | - Lang Lei
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China. .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.
| |
Collapse
|
23
|
Kong S, Zhao YG, Guo L, Gao M, Jin C, She Z. Transcriptomics of Planococcus kocurii O516 reveals the degrading metabolism of sulfamethoxazole in marine aquaculture wastewater. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 265:114939. [PMID: 32540599 DOI: 10.1016/j.envpol.2020.114939] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/18/2020] [Accepted: 06/01/2020] [Indexed: 06/11/2023]
Abstract
Environmental threat induced by residual antibiotics in marine aquaculture wastewater is an urgent problem to be solved. In this study, one sulfamethoxazole (SMX)-degrading bacterium, Planococcus kocurii O516 was isolated from high SMX marine aquafarm. The isolate was able to consume more than 60% of SMX with the initial concentration of 10 mg L-1 within 72 h. Transcriptome analysis found great gene expression differences in the strains with or without SMX dosage. Three putatively differentially expressed proteins, namely AbrB/MazE/SpoVT family DNA-binding domain-containing protein, pantoate-beta-alanine ligase and MerR family transcriptional regulator, were annotated in detail. They were inferred to trigger the strain's response to SMX stress. Reverse transcription-quantitative PCR (RT-qPCR) analysis of four significantly different expressed genes accorded well with expression changes revealed by transcriptomics and confirmed the validity of transcriptome analysis. According to functional annotations of the proteins obtained by transcriptome sequencing and structural analysis of the intermediate metabolites by GC-MS, a possible SMX degradation pathway was reasonably proposed. SMX was first decomposed into sulfonamide and 5-methylisoxazole. The sulfonamide was then hydroxylated to form 4-(hydroxyamino) benzenesulfonamide. Subsequently, the sulfamic acid was detached, and 4-(hydroxyamino) phenol was formed. Finally, 4-aminophenol was generated from dehydroxylated of 4-(hydroxyamino) phenol. In sum, transcriptome analysis of the P. kocurii in response to SMX stress benefits to revealing the degradation pathway of SMX and will provide theoretical feasibility for the application of microbial method to treat the SMX-contaminated aquaculture wastewater.
Collapse
Affiliation(s)
- Sijia Kong
- Shandong Provincial Key Laboratory of Marine Environment and Geological Engineering (MEGE), College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China
| | - Yang-Guo Zhao
- Shandong Provincial Key Laboratory of Marine Environment and Geological Engineering (MEGE), College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China; Key Laboratory of Marine Environment and Ecology (Ocean University of China), Ministry of Education, Qingdao, 266100, China.
| | - Liang Guo
- Shandong Provincial Key Laboratory of Marine Environment and Geological Engineering (MEGE), College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China; Key Laboratory of Marine Environment and Ecology (Ocean University of China), Ministry of Education, Qingdao, 266100, China
| | - Mengchun Gao
- Shandong Provincial Key Laboratory of Marine Environment and Geological Engineering (MEGE), College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China; Key Laboratory of Marine Environment and Ecology (Ocean University of China), Ministry of Education, Qingdao, 266100, China
| | - Chunji Jin
- Shandong Provincial Key Laboratory of Marine Environment and Geological Engineering (MEGE), College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China; Key Laboratory of Marine Environment and Ecology (Ocean University of China), Ministry of Education, Qingdao, 266100, China
| | - Zonglian She
- Shandong Provincial Key Laboratory of Marine Environment and Geological Engineering (MEGE), College of Environmental Science and Engineering, Ocean University of China, Qingdao, 266100, China; Key Laboratory of Marine Environment and Ecology (Ocean University of China), Ministry of Education, Qingdao, 266100, China
| |
Collapse
|
24
|
Abstract
Iron chelators have long been a target of interest as anticancer agents. Iron is an important cellular resource involved in cell replication, metabolism and growth. Iron metabolism is modulated in cancer cells reflecting their increased replicative demands. Originally, iron chelators were first developed for use in iron overload disorders, however, their potential as anticancer agents has been gaining increasing interest. This is due, in part, to the downstream effects of iron depletion such as the inhibition of proliferation through ribonucleotide reductase activity. Additionally, some chelators form redox active metal complexes with iron resulting in the production of reactive oxygen species and oxidative stress. Newer synthetic iron chelators such as Deferasirox, Triapine and di-2-pyridylketone-4,4,-dimethyl-3-thiosemicrbazone (Dp44mt) have improved pharmacokinetic properties over the older chelator Deferoxamine. This review examines and discusses the various iron chelators that have been trialled for cancer therapy including both preclinical and clinical studies. The successes and shortcomings of each of the chelators and their use in combination therapies are highlighted and future potential in the cancer therapy world is considered.
Collapse
|
25
|
Qi J, Zhou N, Li L, Mo S, Zhou Y, Deng Y, Chen T, Shan C, Chen Q, Lu B. Ciclopirox activates PERK-dependent endoplasmic reticulum stress to drive cell death in colorectal cancer. Cell Death Dis 2020; 11:582. [PMID: 32719342 PMCID: PMC7385140 DOI: 10.1038/s41419-020-02779-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 02/08/2023]
Abstract
Ciclopirox (CPX) modulates multiple cellular pathways involved in the growth of a variety of tumor cell types. However, the effects of CPX on colorectal cancer (CRC) and the underlying mechanisms for its antitumor activity remain unclear. Herein, we report that CPX exhibited strong antitumorigenic properties in CRC by inducing cell cycle arrest, repressing cell migration, and invasion by affecting N-cadherin, Snail, E-cadherin, MMP-2, and MMP-9 expression, and disruption of cellular bioenergetics contributed to CPX-associated inhibition of cell growth, migration, and invasion. Interestingly, CPX-induced reactive oxygen species (ROS) production and impaired mitochondrial respiration, whereas the capacity of glycolysis was increased. CPX (20 mg/kg, intraperitoneally) substantially inhibited CRC xenograft growth in vivo. Mechanistic studies revealed that the antitumor activity of CPX relies on apoptosis induced by ROS-mediated endoplasmic reticulum (ER) stress in both 5-FU-sensitive and -resistant CRC cells. Our data reveal a novel mechanism for CPX through the disruption of cellular bioenergetics and activating protein kinase RNA-like endoplasmic reticulum kinase (PERK)-dependent ER stress to drive cell death and overcome drug resistance in CRC, indicating that CPX could potentially be a novel chemotherapeutic for the treatment of CRC.
Collapse
Affiliation(s)
- Jianjun Qi
- Protein Quality Control and Diseases laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.,Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ningning Zhou
- Protein Quality Control and Diseases laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Liyi Li
- Department of Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Shouyong Mo
- Department of Laboratory Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 32300, China
| | - Yidan Zhou
- Protein Quality Control and Diseases laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yao Deng
- Protein Quality Control and Diseases laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ting Chen
- Protein Quality Control and Diseases laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Qin Chen
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Bin Lu
- Protein Quality Control and Diseases laboratory, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
26
|
Wang L, Li X, Mu Y, Lu C, Tang S, Lu K, Qiu X, Wei A, Cheng Y, Wei W. The iron chelator desferrioxamine synergizes with chemotherapy for cancer treatment. J Trace Elem Med Biol 2019; 56:131-138. [PMID: 31466045 DOI: 10.1016/j.jtemb.2019.07.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cisplatin (CDDP) resistance remains a major obstacle for treatment of ovarian cancer. Iron contributes to the growth and reproduction of malignant cells, thus iron chalators can inhibit the growth of tumor cells by depleting the intracellular iron pool. The iron chelator, desferrioxamine (DFO), has performed anticancer in previous study. The aim of our study is to determine the correlation between iron-deprivation and tumor chemosensitivity in ovarian cancer. METHODS To investigate the prognostic value of ferritin light (FTL), ferroportin (FPN), hepcidin (HAMP) and divalent metal-ion transporter-1 (DMT1) in ovarian cancer, the Kaplan-Meier analysis and the Gene Expression Profiling Interactive Analysis (GEPIA) were used. The ovarian cancer cell lines (SKOV-3 and OVCAR-3) were exposed to a gradient concentration of DFO (10, 20, 50, 100, 200 μM) and CDDP (1, 5, 10, 50,100 μM) for 24 h. The protein expression of FTL was tested. The expression of cancer stem cell (CSC) markers, including Sox2, Nanog and C-myc, were downregulated with treatment of DFO. Also, the mamosphere formation and the plation of CD44+/high/CD133+/high and Aldehyde dehydrogenase (ALDH)+/high SKOV-3 cells were reduced after treatment for 7d. Furthermore, we detected the expression of p53, BCL-2, BAX, and caspase-8. RESULTS The survival analysis revealed that high expression of FTL, DMT1, HAMP, showed poor overall survival (OS) in ovarian cancer patients. Our combined data found that DFO could effectively inhibit CSCs, improve the resistance to chemotherapy, and significantly enhanced the efficacy of CDDP therapy in vitro in promoting apoptosis. Besides, targeting molecular targets, including BAX, BCL-2, p53 and caspase-8 could serve as the clinical biomarkers to evaluate the effects of ovarian cancer. It is reasonable to believe that DFO adjuvant therapy in combination with CDDP chemotherapy can promote the improvement of treatment response in ovarian cancer patients. CONCLUSION Our research suggests the experimental evidence for DFO and CDDP as a new effective combination therapy to enhance the efficacy of chemical therapy in ovarian cancer.
Collapse
Affiliation(s)
- Lingjuan Wang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China; Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xiaoqing Li
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China
| | - Yanxi Mu
- Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, 510220, China
| | - Chang Lu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China
| | - Shiqian Tang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China
| | - Kun Lu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiaoming Qiu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China
| | - Aili Wei
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China
| | - Yongjiu Cheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Wei Wei
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
27
|
Yu L, Chen W, Tang Q, Ji KY. Micheliolide Inhibits Liver Cancer Cell Growth Via Inducing Apoptosis And Perturbing Actin Cytoskeleton. Cancer Manag Res 2019; 11:9203-9212. [PMID: 31754310 PMCID: PMC6825479 DOI: 10.2147/cmar.s216870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Micheliolide (MCL) is an effector compound of the flower which has been traditionally used to treat inflammation and cancer patients in oriental medicine. MCL has killing effects on several cancer and immune cells by modulating apoptosis, cell cycle, and metabolism. However, the detail of the mechanisms of anti-cancer activity remains to be elucidated and the effect on liver cancer cells is unknown. Methods Cell proliferation was determined by CCK8 and clone formation assay. The xenograft liver cancer model formed by injecting Huh7 cells into NUDE mice was used to evaluate the effects of MCL on liver cancer cells in vivo. We evaluated the stemness of cells with spheroid formation assay and flow cytometry assay. The apoptosis was determined by Annexin V assay. F-actin staining and ROS were performed to detect the impairment of the F-actin cytoskeleton and mitochondria. Results Here, we first show that MCL inhibits liver cancer cells both in vivo and in vitro by triggering apoptosis which was reduced by anti-oxidant, but not cell-cycle arrest. In addition, MCL induces mitochondrial ROS and caspase-3 activation. Also, we found that the aggregation of mitochondria and the perturbation of F-actin fibers in the MCL-treated liver cancer cells coincidently occurred before the induction of apoptosis and mitochondrial ROS. Conclusion These results suggest that F-actin perturbation is involved in impaired mitochondria and apoptosis. Therefore, MCL can be a potent therapeutic reagent for liver cancer, primarily targeting the actin cytoskeleton.
Collapse
Affiliation(s)
- Lili Yu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, People's Republic of China.,Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Wancheng Chen
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Qingshuang Tang
- Cancer Research Institute, Guangdong Provincial Key Laboratory of Cancer Immunotherapy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Kai-Yuan Ji
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, People's Republic of China
| |
Collapse
|
28
|
Li Y, Lu J, Chen Q, Han S, Shao H, Chen P, Jin Q, Yang M, Shangguan F, Fei M, Wang L, Liu Y, Liu N, Lu B. Artemisinin suppresses hepatocellular carcinoma cell growth, migration and invasion by targeting cellular bioenergetics and Hippo-YAP signaling. Arch Toxicol 2019; 93:3367-3383. [PMID: 31563988 DOI: 10.1007/s00204-019-02579-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/17/2019] [Indexed: 01/17/2023]
Abstract
The primary liver cancer (PLC) is one of the leading causes of cancer-related death worldwide. The predominant form of PLC is hepatocellular carcinoma (HCC), which accounts for about 85% of all PLC. Artemisinin (ART) was clinically used as anti-malarial agents. Recently, it was demonstrated to inhibit cell growth and migration in multiple cancer types. However, the molecular mechanism underlying these anti-cancer activity remains largely unknown. Herein, it is discovered that ART dramatically suppresses HCC cell growth in vitro through arresting cell cycle progression, and represses cell migration and invasion via regulating N-cadherin-Snail-E-cadherin axis. In addition, the disruption of cellular bioenergetics contributed to ART-caused cell growth, migration and invasion inhibition. Moreover, ART (100 mg/kg, intraperitoneally) substantially inhibits HCC xenograft growth in vivo. Importantly, Hippo-YAP signal transduction is remarkably inactivated in HCC cells upon ART administration. Collectively, these data reveal a novel mechanism of ART in regulating HCC cell growth, migration, and invasion, which indicates that ART could be considered as a potential drug for the treatment of HCC.
Collapse
Affiliation(s)
- Yujie Li
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China.,Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jing Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Qin Chen
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Shengnan Han
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Hua Shao
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Pingyi Chen
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Qiumei Jin
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Mingyue Yang
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Fugen Shangguan
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Mingming Fei
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Lu Wang
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Yongzhang Liu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China. .,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Naxin Liu
- Department of Pancreatitis Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China. .,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Bin Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China. .,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
29
|
Zhang J, Hu W, Ding C, Yao G, Zhao H, Wu S. Deferoxamine inhibits iron-uptake stimulated osteoclast differentiation by suppressing electron transport chain and MAPKs signaling. Toxicol Lett 2019; 313:50-59. [PMID: 31238089 DOI: 10.1016/j.toxlet.2019.06.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022]
Abstract
Iron overload causes osteoporosis by enhancing osteoclastic bone resorption. During differentiation, osteoclasts demand high energy and contain abundant mitochondria. In mitochondria, iron is used for the synthesis of Fe-S clusters to support mitochondria biogenesis and electron transport chain. Moreover, mitochondrial reactive oxygen species (ROS) play an important role in osteoclastogenesis. Activation of MAPKs (ERK1/2, JNK, and p38) by ROS is essential and contribute to osteoclast differentiation. How iron chelation impairs electron transport chain and ROS dependent MAPKs activation during osteoclast differentiation is unknown. This study aimed to determine the direct effects of iron chelation on osteoclast differentiation, electron transport chain and MAPKs activation. In the present study, we found that when iron chelator, deferoxamine (DFO), was added, a dose-dependent inhibition of osteoclast differentiation and bone resorption was observed. Supplementation of transferrin-bound iron recovered osteoclastogenesis. Iron chelation resulted in a marked decrease in ferritin level, and increased expression of transferrin receptor 1 and ferroportin. As an iron chelator, DFO negatively affected mitochondrial function through decreasing activities of all the complexes. Expressions of mitochondrial subunits encoded both by mitochondrial and nuclear DNA were decreased. DFO augmented production of mitochondrial ROS, but inhibited the phosphorylation of ERK1/2, JNK, and p38, even in the presence of hydrogen peroxide. These results suggest that iron chelation directly inhibits iron-uptake stimulated osteoclast differentiation and suppresses electron transport chain. Iron chelation negatively regulates MAPKs activation, and this negative regulation is independent on ROS stimulation.
Collapse
Affiliation(s)
- Jian Zhang
- Institute of Laboratory Animal Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China.
| | - Wentao Hu
- School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Chong Ding
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Department of Biomedical Engineering, School of Electrical Engineering, Hebei University of Technology, Tianjin, China
| | - Gang Yao
- Institute of Laboratory Animal Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hai Zhao
- Institute of Laboratory Animal Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Shuguang Wu
- Institute of Laboratory Animal Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
30
|
Zhang J, Sun L, Li W, Wang Y, Li X, Liu Y. Overexpression of macrophage stimulating 1 enhances the anti-tumor effects of IL-24 in esophageal cancer via inhibiting ERK-Mfn2 signaling-dependent mitophagy. Biomed Pharmacother 2019; 114:108844. [PMID: 30981108 DOI: 10.1016/j.biopha.2019.108844] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Although cytokine-based therapy is a promising tool to control the progression of esophageal cancer, low therapeutic responses largely compromise treatment efficacy through unidentified mechanisms. The goal of our study was to explore the roles of macrophage stimulating 1 (Mst1) and mitophagy in enhancing IL-24-based cytokine therapy in esophageal cancer. Our data demonstrated that IL-24 application promoted cancer death by inducing mitochondrial stress, as manifested by mitochondrial ROS overproduction, mitochondrial potential dissipation, cellular ATP deprivation and mitochondrial death activation. Overexpression of Mst1 enhanced IL-24-mediated mitochondrial damage and further augmented IL-24-induced death in esophageal cancer. Molecular investigations illustrated that the IL-24-activated mitochondrial response is accompanied by activation of mitophagy, a protective mechanism to attenuate mitochondrial damage. However, Mst1 overexpression inhibited mitophagy activity, which was achieved by inactivating the ERK-Mfn2 signaling pathway. The re-activation of mitophagy abolished the cancer-killing effects of Mst1 overexpression on esophageal cancer. Altogether, our data demonstrate that IL-24-related therapeutic resistance is associated with mitophagy activation. Mst1 overexpression inhibits mitophagy activity via suppressing the ERK-Mfn2 pathway, ultimately augmenting IL-24-inducd esophageal cancer death via enhanced mitochondrial stress.
Collapse
Affiliation(s)
- Jianpeng Zhang
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Lin Sun
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Weiqiang Li
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Yanyu Wang
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Xinzhen Li
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| | - Yang Liu
- Department of Thoracic Surgery, Beijing Luhe Hospital, Capital Medical University, 82 Xinhua South Road, Tongzhou District, Beijing, 101149, PR China.
| |
Collapse
|
31
|
Flavin Oxidase-Induced ROS Generation Modulates PKC Biphasic Effect of Resveratrol on Endothelial Cell Survival. Biomolecules 2019; 9:biom9060209. [PMID: 31151226 PMCID: PMC6628153 DOI: 10.3390/biom9060209] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Dietary intake of natural antioxidants is thought to impart protection against oxidative-associated cardiovascular diseases. Despite many in vivo studies and clinical trials, this issue has not been conclusively resolved. Resveratrol (RES) is one of the most extensively studied dietary polyphenolic antioxidants. Paradoxically, we have previously demonstrated that high RES concentrations exert a pro-oxidant effect eventually elevating ROS levels leading to cell death. Here, we further elucidate the molecular determinants underpinning RES-induced oxidative cell death. Methods: Using human umbilical vein endothelial cells (HUVECs), the effect of increasing concentrations of RES on DNA synthesis and apoptosis was studied. In addition, mRNA and protein levels of cell survival or apoptosis genes, as well as protein kinase C (PKC) activity were determined. Results: While high concentrations of RES reduce PKC activity, inhibit DNA synthesis and induce apoptosis, low RES concentrations elicit an opposite effect. This biphasic concentration-dependent effect (BCDE) of RES on PKC activity is mirrored at the molecular level. Indeed, high RES concentrations upregulate the proapoptotic Bax, while downregulating the antiapoptotic Bcl-2, at both mRNA and protein levels. Similarly, high RES concentrations downregulate the cell cycle progression genes, c-myc, ornithine decarboxylase (ODC) and cyclin D1 protein levels, while low RES concentrations display an increasing trend. The BCDE of RES on PKC activity is abrogated by the ROS scavenger Tempol, indicating that this enzyme acts downstream of the RES-elicited ROS signaling. The RES-induced BCDE on HUVEC cell cycle machinery was also blunted by the flavin inhibitor diphenyleneiodonium (DPI), implicating flavin oxidase-generated ROS as the mechanistic link in the cellular response to different RES concentrations. Finally, PKC inhibition abrogates the BCDE elicited by RES on both cell cycle progression and pro-apoptotic gene expression in HUVECs, mechanistically implicating PKC in the cellular response to different RES concentrations. Conclusions: Our results provide new molecular insight into the impact of RES on endothelial function/dysfunction, further confirming that obtaining an optimal benefit of RES is concentration-dependent. Importantly, the BCDE of RES could explain why other studies failed to establish the cardio-protective effects mediated by natural antioxidants, thus providing a guide for future investigation looking at cardio-protection by natural antioxidants.
Collapse
|
32
|
Li B, Espósito BP, Wang S, Zhang J, Xu M, Zhang S, Zhang Z, Liu S. Desferrioxamine-caffeine shows improved efficacy in chelating iron and depleting cancer stem cells. J Trace Elem Med Biol 2019; 52:232-238. [PMID: 30732888 DOI: 10.1016/j.jtemb.2019.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/04/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022]
Abstract
Iron chelation has already been proposed to be a feasible strategy for cancer therapeutics in that reinforced iron demand is demonstrated in cancer cells, and quite a few iron chelators have been developed for this purpose. Desferrioxamine (DFO), an iron chelator approved by the U.S. Food and Drug Administration (FDA), has been extensively examined to remove extra iron. However, DFO has been found to harbor limited efficacies in combating cancer cells due to poor cellular permeability. In the current study, we synthesized the DFO derivative, named as desferrioxamine-caffeine dimer (DFCAF) by linking DFO to caffeine with high purity and excellent stability. Our data showed that DFCAF displayed greater cellular permeability to chelate intracellular iron in 4T1 breast cancer cells than DFO, posing more inhibition on cell growth and cellular motility/invasion. Importantly, DFCAF was uncovered to remarkably deplete cancer stem cells (CSCs), as characterized by the remarkable decrease of the CD44+/high/CD24-/low and ALDH+/high subpopulation. In parallel, DFCAF was also found to greatly reverse epithelial-mesenchymal transition (EMT), suggesting the potential application to restrain tumor progression and metastasis. Collectively, these data unveiled the improved efficacy to target cancer cells and to deplete CSCs, thus opening a new path for better cancer therapeutics through iron chelation.
Collapse
Affiliation(s)
- Bin Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital, Tianjin Medical University, Tianjin 300211, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Breno Pannia Espósito
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, Brazil
| | - Shunhao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuping Zhang
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zhihong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital, Tianjin Medical University, Tianjin 300211, China.
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|