1
|
Shi J, Han W, Wang J, Kong X. Anti-Tumor Strategies Targeting Nutritional Deprivation: Challenges and Opportunities. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2415550. [PMID: 39895165 DOI: 10.1002/adma.202415550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/04/2025] [Indexed: 02/04/2025]
Abstract
Higher and richer nutrient requirements are typical features that distinguish tumor cells from AU: cells, ensuring adequate substrates and energy sources for tumor cell proliferation and migration. Therefore, nutrient deprivation strategies based on targeted technologies can induce impaired cell viability in tumor cells, which are more sensitive than normal cells. In this review, nutrients that are required by tumor cells and related metabolic pathways are introduced, and anti-tumor strategies developed to target nutrient deprivation are described. In addition to tumor cells, the nutritional and metabolic characteristics of other cells in the tumor microenvironment (including macrophages, neutrophils, natural killer cells, T cells, and cancer-associated fibroblasts) and related new anti-tumor strategies are also summarized. In conclusion, recent advances in anti-tumor strategies targeting nutrient blockade are reviewed, and the challenges and prospects of these anti-tumor strategies are discussed, which are of theoretical significance for optimizing the clinical application of tumor nutrition deprivation strategies.
Collapse
Affiliation(s)
- Jinsheng Shi
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Wei Han
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Jie Wang
- Pharmacy Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, Shandong, 266000, China
| | - Xiaoying Kong
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong, 266071, China
| |
Collapse
|
2
|
Zhang R, Shen Y, Zhou X, Li J, Zhao H, Zhang Z, Zhao J, Jin H, Guo S, Ding H, Nie G, Zhang Z, Wang Y, Yan X, Fan K. Hypoxia-tropic delivery of nanozymes targeting transferrin receptor 1 for nasopharyngeal carcinoma radiotherapy sensitization. Nat Commun 2025; 16:890. [PMID: 39837820 PMCID: PMC11751138 DOI: 10.1038/s41467-025-56134-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC), a malignancy highly prevalent in East and Southeast Asia, is primarily treated with radiotherapy (RT). However, hypoxia-induced radioresistance presents a significant challenge. Nanozymes, nanomaterials with catalase-like activity, have emerged as a promising strategy for radiosensitization by converting elevated hydrogen peroxide in the tumor microenvironment into oxygen. Despite their potential, effectively targeting hypoxic lesions has been difficult. Here, we identify transferrin receptor 1 (TfR1) as an upregulated target in NPC, with its expression levels positively correlated with hypoxia. Human heavy-chain ferritin, a specific ligand of TfR1, selectively recognizes hypoxic NPC lesions in preclinical models. Based on these findings, we design a hypoxia-targeted nanozyme by loading platinum nanoparticles into ferritin. This nanozyme exhibits enhanced catalase-like activity and effectively alleviates tumor hypoxia in NPC xenografts. When combined with RT, a single injection of the nanozyme significantly inhibits tumor growth and prolongs mouse survival, outperforming sodium glycididazole, a clinically used radiosensitizer. In summary, our findings highlight TfR1 as an accessible cell surface target in hypoxic NPC lesions. The nanozyme targeting TfR1 holds promise for enhancing the therapeutic effectiveness of RT in NPC through an in situ oxygen-generation mechanism.
Collapse
Affiliation(s)
- Ruofei Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yanfang Shen
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiaoying Zhou
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning, China
| | - Jianru Li
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Hanqing Zhao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Zixia Zhang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Jun Zhao
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Ministry of Education, Nanning, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Shuanshuan Guo
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Hui Ding
- Shenzhen Key Laboratory of nanozymes and Translational Cancer Research, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Guohui Nie
- Shenzhen Key Laboratory of nanozymes and Translational Cancer Research, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhe Zhang
- Department of Otolaryngology-Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Ying Wang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China.
| |
Collapse
|
3
|
Wang Z, Xiang S, Qiu Y, Yu F, Li S, Zhang S, Song G, Xu Y, Meng T, Yuan H, Hu F. An "Iron-phagy" nanoparticle inducing irreversible mitochondrial damages for antitumor therapy. J Control Release 2024; 374:400-414. [PMID: 39153721 DOI: 10.1016/j.jconrel.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Cellular iron is inseparably related with the proper functionalities of mitochondria for its potential to readily donate and accept electrons. Though promising, the available endeavors of iron chelation antitumor therapies have tended to be adjuvant therapies. Herein, we conceptualized and fabricated an "iron-phagy" nanoparticle (Dp44mT@HTH) capable of inducing the absolute devastation of mitochondria via inhibiting the autophagy-removal of impaired ones for promoting cancer cell death. The Dp44mT@HTH with hyaluronic acid (HA) as hydrophilic shell can specifically target the highly expressed CD44 receptors on the surface of 4T1 tumor cells. After internalization and lysosomal escape, the nanoparticle disassembles in response to the reactive oxygen species (ROS), subsequently releasing the iron chelator Dp44mT and autophagy-inhibitory drug hydroxychloroquine (HCQ). Dp44mT can then seize cellular Fe2+ to trigger mitochondrial dysfunction via respiratory chain disturbance, while HCQ not only lessens Fe2+ intake, but also impedes fusions of autophagosomes and lysosomes. Consequentially, Dp44mT@HTH induces irreversible mitochondrial impairments, in this respect creating a substantial toxic stack state that induces apoptosis and cell death. Initiating from the perspective of endogenous substances, this strategy illuminates the promise of iron depletion therapy via irreversible mitochondrial damage induction for anticancer treatment.
Collapse
Affiliation(s)
- Zixu Wang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shanshan Xiang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yihe Qiu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Fangying Yu
- Department of Diagnostic Ultrasound and Echocardiography, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Sufen Li
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shufen Zhang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Guangtao Song
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yichong Xu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
4
|
Lin H, Lin S, Shi L, Xu G, Lin M, Li S, Chen J, Li Z, Nakazibwe C, Xiao Y, Li X, Pan X, Wang C. FGFR1 governs iron homeostasis via regulating intracellular protein degradation pathways of IRP2 in prostate cancer cells. Commun Biol 2024; 7:1011. [PMID: 39154074 PMCID: PMC11330447 DOI: 10.1038/s42003-024-06704-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024] Open
Abstract
The acquisition of ectopic fibroblast growth factor receptor 1 (FGFR1) expression is well documented in prostate cancer (PCa) progression, notably in conferring tumor growth advantage and facilitating metastasis. However, how FGFR1 contributes to PCa progression is not fully revealed. Here we report that ectopic FGFR1 in PCa cells promotes transferrin receptor 1 (TFR1) expression and expands the labile iron pool (LIP), and vice versa. We further demonstrate that FGFR1 stabilizes iron regulatory proteins 2 (IRP2) and therefore, upregulates TFR1 via promoting IRP2 binding to the IRE of TFR1. Deletion of FGFR1 in DU145 cells decreases the LIP, which potentiates the anticancer efficacy of iron chelator. Intriguingly, forced expression of IRP2 in FGFR1 depleted cells reinstates TFR1 expression and LIP, subsequently restoring the tumorigenicity of the cells. Together, our results here unravel a new mechanism by which FGFR1 drives PCa progression and suggest a potential novel target for PCa therapy.
Collapse
Affiliation(s)
- Hui Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuaijun Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liuhong Shi
- Department of Head and Neck Surgery, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guangsen Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Manjie Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Supeng Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiale Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiquan Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Catherine Nakazibwe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunbei Xiao
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xuebo Pan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Cong Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
5
|
Zhang WJ, Hu CL, Guo BL, Liang XP, Wang CY, Yang T. STAT5B Suppresses Ferroptosis by Promoting DCAF13 Transcription to Regulate p53/xCT Pathway to Promote Mantle Cell Lymphoma Progression. Biologics 2024; 18:181-193. [PMID: 38979130 PMCID: PMC11229983 DOI: 10.2147/btt.s461287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
Objective The purpose of this study was to analyze the mechanism by which STAT5B inhibits ferroptosis in mantle cell lymphoma (MCL) by promoting DCAF13 transcriptional regulation of p53/xCT pathway. Methods The correlations between STAT5B, DCAF13 and ferroptosis in MCL were analyzed using Gene Expression Profiling Interactive Analysis (GEPIA, http://gepia.cancer-pku.cn/index.html). The expression levels and pairwise correlations of STAT5B, DCAF13, p53 and xCT in MCL patients were detected, respectively. STAT5B was silenced to confirm their criticality in MCL ferroptosis. the effects of blocking necrosis, apoptosis and ferroptosis on the anti-MCL effects of STAT5B were examined. Cells with STAT5B overexpression and/or DCAF13 silencing were constructed to confirm the involvement of DCAF13 in the STAT5B-regulated p53/xCT pathway. The regulation of p53 ubiquitination was confirmed by DCAF13 overexpression and MG132. The effects of silencing DCAF13 and MG132 on STAT5B overexpression on MCL was clarified by a tumor-bearing nude mouse model. Results DCAF13 was overexpressed in MCL and positively correlated with STAT5B, negatively correlated with p53, and positively correlated with xCT. Inhibition of ferroptosis alleviated the inhibitory effects of siSTAT5B on MCL, while inhibition of necrosis and apoptosis had few effects. Silencing of DCAF13 led to the blocking of STAT5B regulation of p53/xCT and ferroptosis. The changes in DCAF13 and the addition of MG132 did not have statistically significant effects on p53 mRNA. Elevation of DCAF13 resulted in downregulation of p53 protein levels, and this inhibition was reversed by MG132. In animal models, the promotion of MCL and the inhibition of ferroptosis by STAT5B. Silencing of DCAF13 blocked STAT5B inhibition of p53 and induction of xCT, GPX4, and GSH. Conclusion STAT5B suppresses ferroptosis by promoting DCAF13 transcription to regulate p53/xCT pathway to promote MCL progression.
Collapse
Affiliation(s)
- Wen Jun Zhang
- Department of Hematology Oncology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Chong Ling Hu
- Department of Hematology Oncology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Bing Ling Guo
- Department of Hematology Oncology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Xi Ping Liang
- Department of Hematology Oncology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Chao Yu Wang
- Department of Hematology Oncology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Tao Yang
- Department of Hematology Oncology, Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| |
Collapse
|
6
|
Guo H, Mi P. Polymer-drug and polymer-protein conjugated nanocarriers: Design, drug delivery, imaging, therapy, and clinical applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1988. [PMID: 39109479 DOI: 10.1002/wnan.1988] [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: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 01/06/2025]
Abstract
Polymer-drug conjugates and polymer-protein conjugates have been pivotal in the realm of drug delivery systems for over half a century. These polymeric drugs are characterized by the conjugation of therapeutic molecules or functional moieties to polymers, enabling a range of benefits including extended circulation times, targeted delivery, controlled release, and decreased immunogenicity. This review delves into recent advancements and challenges in the clinical translations and preclinical studies of polymer-drug conjugates and polymer-protein conjugates. The design principles and functionalization strategies crucial for the development of these polymeric drugs were explored followed by the review of structural properties and characteristics of various polymer carriers. This review also identifies significant obstacles in the clinical translation of polymer-drug conjugates and provides insights into the directions for their future development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Haochen Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Jin X, Shi X, Zhang T, Li X, Xie Y, Tian S, Han K. MALDI-mass spectrometry imaging as a new technique for detecting non-heme iron in peripheral tissues via caudal vein injection of deferoxamine. Anal Bioanal Chem 2024; 416:3389-3399. [PMID: 38632130 DOI: 10.1007/s00216-024-05289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024]
Abstract
As one of the most common iron-chelating agents, deferoxamine (DFO) rapidly chelates iron in the body. Moreover, it does not compete for the iron characteristic of hemoglobin in the blood cells, which is common in the clinical treatment of iron poisoning. Iron is a trace element necessary to maintain organism normal life activities. Iron deficiency can lead to anemia, whereas iron overload can cause elevated levels of cellular oxidative stress and cell damage. As a consequence, detection of the iron content in tissues and blood is of great significance. The traditional techniques for detecting the iron content include inductively coupled plasma-mass spectrometry and atomic absorption spectrometry, which cannot be used for imaging purposes. Laser ablation-ICP-MS and synchrotron radiation micro-X-ray fluorescence can map the concentration and distribution of iron in tissues. However, these methods can only be used to measure the total iron levels in blood or tissues. In recent years, due to the deepening understanding of iron metabolism, diseases related to iron overload have attracted increasing attention. Therefore, we took advantage of the properties of DFO in terms of chelating iron and investigated different sampling times following DFO injection in the tail vein of mice. We used mass spectrometry imaging (MSI) technology to detect the DFO and ferrioxamine content in the blood and different tissues to indirectly characterize the non-heme iron content.
Collapse
Affiliation(s)
- Xiaofang Jin
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China
| | - Xintong Shi
- College of Chemistry and Materials Science, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China
| | - Tong Zhang
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China
| | - Xingyao Li
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China
| | - Yajing Xie
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China
| | - Siyu Tian
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China
| | - Kang Han
- Analysis and Testing Centre, Hebei Normal University, Shijiazhuang, 050024, Hebei Province, China.
| |
Collapse
|
8
|
Wang B, Kong W, Lv L, Wang Z. Plumbagin induces ferroptosis in colon cancer cells by regulating p53-related SLC7A11 expression. Heliyon 2024; 10:e28364. [PMID: 38596137 PMCID: PMC11002553 DOI: 10.1016/j.heliyon.2024.e28364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Objective This study examined the mechanism through which plumbagin induces ferroptosis of colon cancer cells. Methods CCK-8 assay was performed to examine the viability of colon cancer cells (SW480 and HCT116 cells) after they were treated with 0-, 5-, 10-, 15- and 20-μmol/L plumbagin. Colony formation assay and Transwell assay were used to examine the effects of 15-μmol/L plumbagin on the proliferation, invasive ability. The ferroptosis of SW480 and HCT116 cells and the expression of p-p53, p53 and SLC7A11 were analysed. The effects of blocking necrosis, apoptosis and ferroptosis on the anti-cancer effects of plumbagin were examined. After p53 was silenced, the effects of plumbagin on proliferation, invasion, ferroptosis and SLC7A11 expression were assessed. A tumour-bearing nude mouse model was used to examine the effects of p53 silencing and/or plumbagin on tumour growth, ferroptosis and SLC7A11 expression. Results Plumbagin inhibited the proliferation of SW480 and HCT116 cells and their invasive and colony-forming abilities. It increased Fe2+ levels but significantly decreased GSH and GPX4 levels. When ferroptosis was inhibited, the effects of plumbagin on colon cancer cells were significantly alleviated. Plumbagin promoted the expression and phosphorylation of p53 and inhibited the mRNA and protein levels of SLC7A11. Silencing of p53 counteracted the effects of plumbagin on the ferroptosis and biological behaviour of SW480 and HCT116 cells. In mouse models of colon cancer, silencing of p53 attenuated the tumour-suppressing effects of plumbagin as well as its inhibitory effects on the protein level of SLC7A11 and restored the expression of GSH and GPX4. Conclusion Plumbagin promotes ferroptosis and inhibits cell proliferation and invasion by decreasing the protein expression of SLC7A11 through p53.
Collapse
Affiliation(s)
- Bingyi Wang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqi Kong
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lixin Lv
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiqiang Wang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Raptania CN, Zakia S, Fahira AI, Amalia R. Article review: Brazilin as potential anticancer agent. Front Pharmacol 2024; 15:1355533. [PMID: 38515856 PMCID: PMC10955326 DOI: 10.3389/fphar.2024.1355533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Brazilin is the main compound in Caesalpinia sappan and Haematoxylum braziletto, which is identified as a homoisoflavonoid based on its molecular structure. These plants are traditionally used as an anti-inflammatory to treat fever, hemorrhage, rheumatism, skin problems, diabetes, and cardiovascular diseases. Recently, brazilin has increased its interest in cancer studies. Several findings have shown that brazilin has cytotoxic effects on colorectal cancer, breast cancer, lung cancer, multiple myeloma, osteosarcoma, cervical cancer, bladder carcinoma, also other cancers, along with numerous facts about its possible mechanisms that will be discussed. Besides its flavonoid content, brazilin is able to chelate metal ions. A study has proved that brazilin could be used as an antituberculosis agent based on its ability to chelate iron. This possible iron-chelating of brazilin and all the studies discussed in this review will lead us to the statement that, in the future, brazilin has the potency to be a chemo-preventive and anticancer agent. The article review aimed to determine the brazilin mechanism and pathogenesis of cancer.
Collapse
Affiliation(s)
- Callista Najla Raptania
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Syifa Zakia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Alistia Ilmiah Fahira
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Laboratory of Cell and Molecular Biology, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Riezki Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Laboratory of Cell and Molecular Biology, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
10
|
Arbon D, Mach J, Čadková A, Sipkova A, Stursa J, Klanicová K, Machado M, Ganter M, Levytska V, Sojka D, Truksa J, Werner L, Sutak R. Chelation of Mitochondrial Iron as an Antiparasitic Strategy. ACS Infect Dis 2024; 10:676-687. [PMID: 38287902 PMCID: PMC10862539 DOI: 10.1021/acsinfecdis.3c00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
Iron, as an essential micronutrient, plays a crucial role in host-pathogen interactions. In order to limit the growth of the pathogen, a common strategy of innate immunity includes withdrawing available iron to interfere with the cellular processes of the microorganism. Against that, unicellular parasites have developed powerful strategies to scavenge iron, despite the effort of the host. Iron-sequestering compounds, such as the approved and potent chelator deferoxamine (DFO), are considered a viable option for therapeutic intervention. Since iron is heavily utilized in the mitochondrion, targeting iron chelators in this organelle could constitute an effective therapeutic strategy. This work presents mitochondrially targeted DFO, mitoDFO, as a candidate against a range of unicellular parasites with promising in vitro efficiency. Intracellular Leishmania infection can be cleared by this compound, and experimentation with Trypanosoma brucei 427 elucidates its possible mode of action. The compound not only affects iron homeostasis but also alters the physiochemical properties of the inner mitochondrial membrane, resulting in a loss of function. Furthermore, investigating the virulence factors of pathogenic yeasts confirms that mitoDFO is a viable candidate for therapeutic intervention against a wide spectrum of microbe-associated diseases.
Collapse
Affiliation(s)
- Dominik Arbon
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Jan Mach
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Aneta Čadková
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Anna Sipkova
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| | - Jan Stursa
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
- Laboratory
of Clinical Pathophysiology, Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech
Republic
| | - Kristýna Klanicová
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Prague 128 00, Czech Republic
| | - Marta Machado
- Graduate
Program in Areas of Basic and Applied Biology, Instituto de Ciências
Biomédicas Abel Salazar, Universidade
do Porto, Porto 4050-313, Portugal
- Centre for
Infectious Diseases, Parasitology, Heidelberg
University Hospital, Heidelberg 69120, Germany
| | - Markus Ganter
- Centre for
Infectious Diseases, Parasitology, Heidelberg
University Hospital, Heidelberg 69120, Germany
| | - Viktoriya Levytska
- Institute
of Parasitology, Biology Centre, Academy
of Sciences of the Czech Republic, Branišovská 1160/31, České Budějovice 37005, Czech Republic
| | - Daniel Sojka
- Institute
of Parasitology, Biology Centre, Academy
of Sciences of the Czech Republic, Branišovská 1160/31, České Budějovice 37005, Czech Republic
| | - Jaroslav Truksa
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
| | - Lukáš Werner
- Institute
of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec 25250, Czech Republic
- Laboratory
of Clinical Pathophysiology, Diabetes Centre, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 140 21 Prague, Czech
Republic
| | - Robert Sutak
- Department of Parasitology, Faculty
of Science, Charles University, BIOCEV, Vestec 25250, Czech Republic
| |
Collapse
|
11
|
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
|
12
|
Hu J, Cheng M, Jiang C, Liu L, He Z, Liu L, Yao Y, Li Z, Wang Q. Deferoxamine Mitigates Ferroptosis and Inflammation in Hippocampal Neurons After Subarachnoid Hemorrhage by Activating the Nrf2/TXNRD1 Axis. Mol Neurobiol 2024; 61:1044-1060. [PMID: 37676391 DOI: 10.1007/s12035-023-03525-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/17/2023] [Indexed: 09/08/2023]
Abstract
Ferroptosis is a distinct peroxidation-driven form of cell death tightly involved in subarachnoid hemorrhage (SAH). This study delved into the mechanism of deferoxamine (DFO, an iron chelator) in SAH-induced ferroptosis and inflammation. SAH mouse models were established by endovascular perforation method and injected intraperitoneally with DFO, or intraventricularly injected with the Nrf2 pathway inhibitor ML385 before SAH, followed by detection of neurological function, blood-brain barrier (BBB) permeability, and brain water content. Apoptotic level of hippocampal neurons, symbolic changes of ferroptosis, and levels of pro-inflammatory cytokines were assessed using TUNEL staining, Western blotting, colorimetry, and ELISA. The localization and expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) were detected. HT22 cells were exposed to Hemin as in vitro SAH models and treated with FIN56 to induce ferroptosis, followed by evaluation of the effects of DFO on FIN56-treated HT22 cells. The regulation of Nrf2 in thioredoxin reductase 1 (TXNRD1) was analyzed by co-immunoprecipitation and Western blotting. Moreover, HT22 cells were treated with DFO and ML385 to identify the role of DFO in the Nrf2/TXNRD1 axis. DFO extenuated brain injury, and ferroptosis and inflammation in hippocampal neurons of SAH mice. Nrf2 localized at the CA1 region of hippocampal neurons, and DFO stimulated nuclear translocation of Nrf2 protein in hippocampal neurons of SAH mice. Additionally, DFO inhibited ferroptosis and inflammatory responses in FIN56-induced HT22 cells. Nrf2 positively regulated TXNRD1 protein expression. Indeed, DFO alleviated FIN56-induced ferroptosis and inflammation via activation of the Nrf2/TXNRD1 axis. DFO alleviated neurological deficits, BBB disruption, brain edema, and brain injury in mice after SAH by inhibiting hippocampal neuron ferroptosis via the Nrf2/TXNRD1 axis. DFO ameliorates SAH-induced ferroptosis and inflammatory responses in hippocampal neurons by activating the Nrf2/TXNRD1 axis.
Collapse
Affiliation(s)
- Junting Hu
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Meixiong Cheng
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Chonggui Jiang
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Ling Liu
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Zongze He
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Lingtong Liu
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Yuanpeng Yao
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Zhili Li
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China.
| | - Qi Wang
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
13
|
Qin Y, Li S, Liang L, Zhao S, Ye F. Rational synthesis of FeNiCo-LDH nanozyme for colorimetric detection of deferoxamine mesylate. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 303:123156. [PMID: 37506456 DOI: 10.1016/j.saa.2023.123156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
The accurate surveillance and sensitive detection of deferoxamine mesylate (DFO) is of great significance to ensure the safety of thalassemia major patients. Herein, we report a new nanozyme-based colorimetric sensor platform for DFO detection. First, a metal-organic framework (ZIF-67) was used as a precursor for the synthesis of FeNiCo-LDH (Layered Double Hydroxide, LDH) via an ion exchange reaction stirring at room temperature. The results of electron microscopy and nitrogen adsorption-desorption showed that FeNiCo-LDH exhibited a 3D hollow and mesopores structure, which supplied more exposed active sites and faster transfer of mass. The as-prepared FeNiCo-LDH showed superior peroxidase-like activity with a low Km and high υmax. It can catalyze the decomposition of H2O2 to generate reactive oxygen species (ROS) and further react with 3,3',5,5'-tetramethylbenzidine (TMB) to form blue oxidized TMB (oxTMB), which has a characteristic absorption at 652 nm. Once DFO was introduced, it can complex with FeNiCo-LDH and inhibit the peroxidase-like activity of FeNiCo-LDH, making the color of oxTMB lighter. The quantitative range of DFO was 0.8-28 μM with a detection limit of 0.71 μM. This established method was applied to the detection of DFO content in urine samples of thalassemia patients, and the spiked recoveries were falling between 97.7% and 109.6%, with a relative standard deviation was less than 5%, providing a promising tool for the clinical medication of thalassemia patients.
Collapse
Affiliation(s)
- Yuan Qin
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Shuishi Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Ling Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Fanggui Ye
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
14
|
Bayanbold K, Singhania M, Fath MA, Searby CC, Stolwijk JM, Henrich JB, Pulliam CF, Schoenfeld JD, Mapuskar KA, Sho S, Caster JM, Allen BG, Buettner GR, Spies M, Goswami PC, Petronek MS, Spitz DR. Depletion of Labile Iron Induces Replication Stress and Enhances Responses to Chemoradiation in Non-Small-Cell Lung Cancer. Antioxidants (Basel) 2023; 12:2005. [PMID: 38001858 PMCID: PMC10669787 DOI: 10.3390/antiox12112005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The intracellular redox-active labile iron pool (LIP) is weakly chelated and available for integration into the iron metalloproteins that are involved in diverse cellular processes, including cancer cell-specific metabolic oxidative stress. Abnormal iron metabolism and elevated LIP levels are linked to the poor survival of lung cancer patients, yet the underlying mechanisms remain unclear. Depletion of the LIP in non-small-cell lung cancer cell lines using the doxycycline-inducible overexpression of the ferritin heavy chain (Ft-H) (H1299 and H292), or treatment with deferoxamine (DFO) (H1299 and A549), inhibited cell growth and decreased clonogenic survival. The Ft-H overexpression-induced inhibition of H1299 and H292 cell growth was also accompanied by a significant delay in transit through the S-phase. In addition, both Ft-H overexpression and DFO in H1299 resulted in increased single- and double-strand DNA breaks, supporting the involvement of replication stress in the response to LIP depletion. The Ft-H and DFO treatment also sensitized H1299 to VE-821, an inhibitor of ataxia telangiectasis and Rad2-related (ATR) kinase, highlighting the potential of LIP depletion, combined with DNA damage response modifiers, to alter lung cancer cell responses. In contrast, only DFO treatment effectively reduced the LIP, clonogenic survival, cell growth, and sensitivity to VE-821 in A549 non-small-cell lung cancer cells. Importantly, the Ft-H and DFO sensitized both H1299 and A549 to chemoradiation in vitro, and Ft-H overexpression increased the efficacy of chemoradiation in vivo in H1299. These results support the hypothesis that the depletion of the LIP can induce genomic instability, cell death, and potentiate therapeutic responses to chemoradiation in NSCLC.
Collapse
Affiliation(s)
- Khaliunaa Bayanbold
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Mekhla Singhania
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Melissa A. Fath
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Charles C. Searby
- University of Iowa Hospitals and Clinics, Department Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Jeffrey M. Stolwijk
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - John B. Henrich
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Casey F. Pulliam
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Joshua D. Schoenfeld
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Kranti A. Mapuskar
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Sei Sho
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Joseph M. Caster
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Bryan G. Allen
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Garry R. Buettner
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Maria Spies
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
- University of Iowa Hospitals and Clinics, Holden Comprehensive Cancer Center, Department of Biochemistry and Molecular Biology, Iowa City, IA 52242, USA
| | - Prabhat C. Goswami
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Michael S. Petronek
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| | - Douglas R. Spitz
- University of Iowa Hospitals and Clinics, Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA (M.A.F.); (G.R.B.)
| |
Collapse
|
15
|
Xia Q, Du Z, Chen M, Zhou X, Bai W, Zheng X, Lin L, Zhao Y, Ding J, Wu Z, Zou H, Wang S, Xu L, Li E, Wu B. A protein complex of LCN2, LOXL2 and MMP9 facilitates tumour metastasis in oesophageal cancer. Mol Oncol 2023; 17:2451-2471. [PMID: 37753805 PMCID: PMC10620126 DOI: 10.1002/1878-0261.13529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 09/28/2023] Open
Abstract
During malignant tumour development, the extracellular matrix (ECM) is usually abnormally regulated. Dysregulated expression of lysyl oxidase-like 2 (LOXL2), matrix metalloproteinase 9 (MMP9) and lipocalin 2 (LCN2) are associated with ECM remodelling. In this study, protein-protein interaction assays indicated that LCN2 and LOXL2 interactions and LCN2 and MMP9 interactions occurred both intracellularly and extracellularly, but interactions between LOXL2 and MMP9 only occurred intracellularly. The LCN2/LOXL2/MMP9 ternary complex promoted migration and invasion of oesophageal squamous cell carcinoma (ESCC) cells, as well as tumour growth and malignant progression in vivo, while the iron chelator deferoxamine mesylate (DFOM) inhibited ESCC tumour growth. Co-overexpression of LCN2, LOXL2 and MMP9 enhanced the ability of tumour cells to degrade fibronectin and Matrigel, increased the formation and extension of filopodia, and promoted the rearrangement of microfilaments through upregulation of profilin 1. In addition, the LCN2/LOXL2/MMP9 ternary complex promoted the expression of testican-1 (SPOCK1), and abnormally activated the FAK/AKT/GSK3β signalling pathway. In summary, the LCN2/LOXL2/MMP9 ternary complex promoted the migration and invasion of cancer cells and malignant tumour progression through multiple mechanisms and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Qiaoxi Xia
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
- Central LaboratoryShantou Central HospitalChina
| | - Zepeng Du
- Central LaboratoryShantou Central HospitalChina
- Department of PathologyShantou Central HospitalChina
| | - Mantong Chen
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Xiao Zhou
- Central LaboratoryShantou Central HospitalChina
| | - Wenjing Bai
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Xiaoqi Zheng
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Ling Lin
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Yan Zhao
- Central LaboratoryShantou Central HospitalChina
| | - Jiyu Ding
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Zhisheng Wu
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Haiying Zou
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Shaohong Wang
- Department of PathologyShantou Central HospitalChina
| | - Liyan Xu
- Institute of Oncologic PathologyShantou University Medical CollegeChina
| | - Enmin Li
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| | - Bingli Wu
- Department of Biochemistry and Molecular BiologyShantou University Medical CollegeChina
| |
Collapse
|
16
|
Passari AK, Ruiz-Villafán B, Cruz-Bautista R, Díaz-Domínguez V, Rodríguez-Sanoja R, Sanchez S. Opportunities and challenges of microbial siderophores in the medical field. Appl Microbiol Biotechnol 2023; 107:6751-6759. [PMID: 37755507 PMCID: PMC10589192 DOI: 10.1007/s00253-023-12742-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/28/2023]
Abstract
Siderophores are low-molecular-weight secondary metabolites that function as iron chelators. Under iron-deficiency conditions, they are produced by a wide variety of microbes, allowing them to increase their iron uptake. The primary function of these compounds is the environmental iron scavenging and its transport into the cytosol. Iron is then reduced to its ferrous form to operate as an enzymatic cofactor for various functions, including respiration, nitrogen fixation, photosynthesis, methanogenesis, and amino acid synthesis. Depending on their functional group, siderophores are classified into hydroxamate, catecholate, phenolate, carboxylate, and mixed types. They have achieved great importance in recent years due to their medical applications as antimicrobial, antimalarial, or anticancer drugs, vaccines, and drug-delivery agents. This review integrates current advances in specific healthcare applications of microbial siderophores, delineating new opportunities and challenges as viable therapies to fight against diseases that represent crucial public health problems in the medical field.Key points• Siderophores are low-molecular-weight secondary metabolites functioning as iron chelators.• The siderophore's properties offer viable options to face diverse clinical problems.• Siderophores are alternatives for the enhancement of antibiotic activities.
Collapse
Affiliation(s)
- Ajit Kumar Passari
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Beatriz Ruiz-Villafán
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Rodrigo Cruz-Bautista
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Valerie Díaz-Domínguez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Sergio Sanchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
17
|
Ghaffari K, Bayat A, Ghasemi A. Combination Effect of Deferoxamine and Arsenic Trioxide on Viability and Vitality of APL Like Cell Line. Ethiop J Health Sci 2023; 33:703-710. [PMID: 38784214 PMCID: PMC11111188 DOI: 10.4314/ejhs.v33i4.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/25/2024] Open
Abstract
Background Arsenic trioxide is an activist agent in the treatment of acute promyelocytic leukemia (APL), which acts alone, but has an adverse effect on patients. Moreover, deferoxamine has antiproliferative activity and induces leukopenia. In order to enhance antileukemic effectiveness and to reduce the dosage of arsenic trioxide, the combination effect of it with deferoxamine (DFO) was evaluated on the APL cell line (NB4). Methods In this experimental study, to investigate the cytotoxic effects of ATO/DFO in acute promyelocytic leukemia, the NB4 cell line (provided by Pasteur Institute of Iran) was treated with different doses and then at 24, 48, and 72 hrs intervals, the percentage of survival, cell count, metabolic activity and apoptosis induction were investigated respectively. Also, hTERT gene expression was analyzed by the RT-PCR method. Results We found that DFO alone and in combination with ATO has cytotoxic and antiproliferative effects, and reduces viability and cell metabolic activity in the NB4 cell line in a dose and time-dependent manner. In addition, this combination causes an increase in apoptosis, up-regulation of Caspase-3, and down-regulation of hTERT genes in cells. Conclusion Combined ATO/ DFO treatment cooperatively decreased the mRNA levels of the hTERT and increased the mRNA levels of Caspase-3 in a time-dependent manner compared to DFO alone.
Collapse
Affiliation(s)
- Kazem Ghaffari
- Department of Basic and Laboratory of Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Ali Bayat
- Department of Basic and Laboratory of Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Ali Ghasemi
- Department of Biochemistry and Hematology, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
18
|
He XY, Fan X, Qu L, Wang X, Jiang L, Sang LJ, Shi CY, Lin S, Yang JC, Yang ZZ, Lei K, Li JH, Ju HQ, Yan Q, Liu J, Wang F, Shao J, Xiong Y, Wang W, Lin A. LncRNA modulates Hippo-YAP signaling to reprogram iron metabolism. Nat Commun 2023; 14:2253. [PMID: 37080959 PMCID: PMC10119135 DOI: 10.1038/s41467-023-37871-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/03/2023] [Indexed: 04/22/2023] Open
Abstract
Iron metabolism dysregulation is tightly associated with cancer development. But the underlying mechanisms remain poorly understood. Increasing evidence has shown that long noncoding RNAs (lncRNAs) participate in various metabolic processes via integrating signaling pathway. In this study, we revealed one iron-triggered lncRNA, one target of YAP, LncRIM (LncRNA Related to Iron Metabolism, also named ZBED5-AS1 and Loc729013), which effectively links the Hippo pathway to iron metabolism and is largely independent on IRP2. Mechanically, LncRIM directly binds NF2 to inhibit NF2-LATS1 interaction, which causes YAP activation and increases intracellular iron level via DMT1 and TFR1. Additionally, LncRIM-NF2 axis mediates cellular iron metabolism dependent on the Hippo pathway. Clinically, high expression of LncRIM correlates with poor patient survival, suggesting its potential use as a biomarker and therapeutic target. Taken together, our study demonstrated a novel mechanism in which LncRIM-NF2 axis facilitates iron-mediated feedback loop to hyperactivate YAP and promote breast cancer development.
Collapse
Affiliation(s)
- Xin-Yu He
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, 310058, Hangzhou, Zhejiang, China
| | - Xiao Fan
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, 310058, Hangzhou, Zhejiang, China
| | - Lei Qu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, 310058, Hangzhou, Zhejiang, China
| | - Xiang Wang
- Department of Central Laboratory, the First People's Hospital of Huzhou, 158 Guangchang Back Road, 313000, Huzhou, Zhejiang, P.R. China
| | - Li Jiang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
| | - Ling-Jie Sang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Cheng-Yu Shi
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Siyi Lin
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jie-Cheng Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Zuo-Zhen Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Kai Lei
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jun-Hong Li
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Huai-Qiang Ju
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 510060, Guangzhou, Guangdong, China
| | - Qingfeng Yan
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jian Liu
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, 310002, Hangzhou, Zhejiang, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 314400, Haining, Zhejiang, China
| | - Fudi Wang
- Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
| | - Jianzhong Shao
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yan Xiong
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, 310000, Hangzhou, Zhejiang, China
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA.
| | - Aifu Lin
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, 310058, Hangzhou, Zhejiang, China.
- International School of Medicine, International Institutes of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, 322000, Yiwu, Zhejiang, China.
- Breast Center of the First Affiliated Hospital, School of Medicine, Zhejiang University, 310003, Hangzhou, Zhejiang, China.
| |
Collapse
|
19
|
Nomoto T, Komoto K, Nagano T, Ishii T, Guo H, Honda Y, Ogura SI, Ishizuka M, Nishiyama N. Polymeric iron chelators for enhancing 5-aminolevulinic acid-induced photodynamic therapy. Cancer Sci 2023; 114:1086-1094. [PMID: 36341512 PMCID: PMC9986068 DOI: 10.1111/cas.15637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
5-Aminolevulinic acid (5-ALA) is an amino acid that can be metabolized into a photosensitizer, protoporphyrin IX (PpIX) selectively in a tumor cell, permitting minimally invasive photodynamic diagnosis/therapy. However, some malignant tumor cells have excess intracellular labile iron and facilitate the conversion of PpIX into heme, which compromises the therapeutic potency of 5-ALA. Here, we examined the potential of chelation of such unfavorable intratumoral labile iron in photodynamic therapy (PDT) with 5-ALA hydrochloride, using polymeric iron chelators that we recently developed. The polymeric iron chelator efficiently inactivated the intracellular labile iron in cultured cancer cells and importantly enhanced the accumulation of PpIX, thereby improving the cytotoxicity upon photoirradiation. Even in in vivo study with subcutaneous tumor models, the polymeric iron chelator augmented the intratumoral accumulation of PpIX and the PDT effect. This study suggests that our polymeric iron chelator could be a tool for boosting the effect of 5-ALA-induced PDT by modulating tumor microenvironment.
Collapse
Affiliation(s)
- Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Kana Komoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | | | | | - Haochen Guo
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuto Honda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Shun-Ichiro Ogura
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | | | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| |
Collapse
|
20
|
Liu P, Wang Q, Li K, Bi B, Wen YF, Qiu MJ, Zhao J, Li BB, Zhang CH, He YL. A DFX-based iron nanochelator for cancer therapy. Front Bioeng Biotechnol 2022; 10:1078137. [DOI: 10.3389/fbioe.2022.1078137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Iron as an essential element, is involved in various cellular functions and maintaining cell viability, cancer cell is more dependent on iron than normal cell due to its chief characteristic of hyper-proliferation. Despite that some of the iron chelators exhibited potent and broad antitumor activity, severe systemic toxicities have limited their clinical application. Polyaminoacids, as both drug-delivery platform and therapeutic agents, have attracted great interests owing to their different medical applications and biocompatibility. Herein, we have developed a novel iron nanochelator PL-DFX, which composed of deferasirox and hyperbranched polylysine. PL-DFX has higher cytotoxicity than DFX and this effect can be partially reversed by Fe2+ supplementation. PL-DFX also inhibited migration and invasion of cancer cells, interfere with iron metabolism, induce phase G1/S arrest and depolarize mitochondria membrane potential. Additionally, the anti-tumor potency of PL-DFX was also supported by organoids derived from clinical specimens. In this study, DFX-based iron nanochelator has provided a promising and prospective strategy for cancer therapy via iron metabolism disruption.
Collapse
|
21
|
Xu L, Guan R, Yu B, Li Y, Liu H, Jiang Y. Fluorene methoxycarbonyl-PEG-deferoxamine conjugates "hitchhike" with albumin in situ for iron overload therapy. Int J Pharm 2022; 625:122136. [PMID: 36029994 DOI: 10.1016/j.ijpharm.2022.122136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022]
Abstract
Although deferoxamine (DFO) has been approved for the treatment the iron overloaded diseases, its clinical application is impeded by very short circulation time and its relating toxicity. In this work, the fluorene methoxycarbonyl (FMOC) for "albumin hitchhiking" was used to prolong the plasma circulation time of DFO and reduce toxicity. The designed FMOC-PEG-DFO conjugates were found to reversible bind to albumin and gradually release DFO in vivo. Herein, the FMOC-PEG1000-DFO conjugates could increase 30 times the blood circulation time of DFO with the improvement of the iron elimination efficacy. Meanwhile, the conjugates markedly reduced the cytotoxicity of DFO. Taken together, the result demonstrated the FMOC-PEG1000-DFO conjugates could be a potential therapeutic choice for iron-overload-related diseases.
Collapse
Affiliation(s)
- Linyi Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Rou Guan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Bohong Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Yicheng Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yiguo Jiang
- Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou 215153, China.
| |
Collapse
|
22
|
Attenuation of tryptophan metabolism by Fe chelators: A hypothesis regarding inhibiting tumor suppressive microenvironments in pancreatic ductal adenocarcinoma. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Abedi M, Rahgozar S. Puzzling Out Iron Complications in Cancer Drug Resistance. Crit Rev Oncol Hematol 2022; 178:103772. [PMID: 35914667 DOI: 10.1016/j.critrevonc.2022.103772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/09/2022] Open
Abstract
Iron metabolism are frequently disrupted in cancer. Patients with cancer are prone to anemia and receive transfusions frequently; the condition which results in iron overload, contributing to serious therapeutic complications. Iron is introduced as a carcinogen that may increase tumor growth. However, investigations regarding its impact on response to chemotherapy, particularly the induction of drug resistance are still limited. Here, iron contribution to cell signaling and various molecular mechanisms underlying iron-mediated drug resistance are described. A dual role of this vital element in cancer treatment is also addressed. On one hand, the need to administer iron chelators to surmount iron overload and improve the sensitivity of tumor cells to chemotherapy is discussed. On the other hand, the necessary application of iron as a therapeutic option by iron-oxide nanoparticles or ferroptosis inducers is explained. Authors hope that this paper can help unravel the clinical complications related to iron in cancer therapy.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
24
|
Huff SE, Winter JM, Dealwis CG. Inhibitors of the Cancer Target Ribonucleotide Reductase, Past and Present. Biomolecules 2022; 12:biom12060815. [PMID: 35740940 PMCID: PMC9221315 DOI: 10.3390/biom12060815] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 01/02/2023] Open
Abstract
Ribonucleotide reductase (RR) is an essential multi-subunit enzyme found in all living organisms; it catalyzes the rate-limiting step in dNTP synthesis, namely, the conversion of ribonucleoside diphosphates to deoxyribonucleoside diphosphates. As expression levels of human RR (hRR) are high during cell replication, hRR has long been considered an attractive drug target for a range of proliferative diseases, including cancer. While there are many excellent reviews regarding the structure, function, and clinical importance of hRR, recent years have seen an increase in novel approaches to inhibiting hRR that merit an updated discussion of the existing inhibitors and strategies to target this enzyme. In this review, we discuss the mechanisms and clinical applications of classic nucleoside analog inhibitors of hRRM1 (large catalytic subunit), including gemcitabine and clofarabine, as well as inhibitors of the hRRM2 (free radical housing small subunit), including triapine and hydroxyurea. Additionally, we discuss novel approaches to targeting RR and the discovery of new classes of hRR inhibitors.
Collapse
Affiliation(s)
- Sarah E. Huff
- Department of Pediatrics, University of California, San Diego, CA 92093, USA;
| | - Jordan M. Winter
- Department of Surgery, Division of Surgical Oncology, University Hospitals Cleveland Medical Center, Akron, OH 44106, USA;
| | - Chris G. Dealwis
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence:
| |
Collapse
|
25
|
Liu K, Huang J, Liu J, Li C, Kroemer G, Tang D, Kang R. HSP90 Mediates IFNγ-Induced Adaptive Resistance to Anti-PD-1 Immunotherapy. Cancer Res 2022; 82:2003-2018. [PMID: 35247909 DOI: 10.1158/0008-5472.can-21-3917] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/01/2022] [Accepted: 03/02/2022] [Indexed: 01/04/2023]
Abstract
SIGNIFICANCE This study reveals an HSP90-centric, iron-modulated mechanism that confers immunosuppression, offering potential therapeutic targets for interfering with acquired resistance to the most prevalent anticancer immunotherapies.
Collapse
Affiliation(s)
- Ke Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Huang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
26
|
Iron Chelators in Treatment of Iron Overload. J Toxicol 2022; 2022:4911205. [PMID: 35571382 PMCID: PMC9098311 DOI: 10.1155/2022/4911205] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 01/19/2023] Open
Abstract
Patients suffering from iron overload can experience serious complications. In such patients, various organs, such as endocrine glands and liver, can be damaged. Although iron is a crucial element for life, iron overload can be potentially toxic for human cells due to its role in generating free radicals. In the past few decades, there has been a major improvement in the survival of patients who suffer from iron overload due to the application of iron chelation therapy in clinical practice. In clinical use, deferoxamine, deferiprone, and deferasirox are the three United States Food and Drug Administration-approved iron chelators. Each of these iron chelators is well known for the treatment of iron overload in various clinical conditions. Based on several up-to-date studies, this study explained iron overload and its clinical symptoms, introduced each of the above-mentioned iron chelators, and evaluated their advantages and disadvantages with an emphasis on combination therapy, which in recent studies seems a promising approach. In numerous clinical conditions, due to the lack of accurate indicators, choosing a standard approach for iron chelation therapy can be difficult; therefore, further studies on the issue are still required. This study aimed to introduce each of these iron chelators, combination therapy, usage doses, specific clinical applications, and their advantages, toxicity, and side effects.
Collapse
|
27
|
Complementary and Alternative Therapies in Oncology. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095071. [PMID: 35564468 PMCID: PMC9104744 DOI: 10.3390/ijerph19095071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022]
Abstract
Cancer is the second leading cause of death worldwide, after cardiovascular diseases. Increasing patients’ awareness and providing easier access to public information result in greater interest in alternative anticancer or unproven supportive therapies. Fear of cancer and limited trust in the treating physician are also important reasons leading patients to seek these methods. Trust and good communication are essential to achieving truthful collaboration between physicians and patients. Given the popularity of CAM, better knowledge about these alternative practices may help oncologists discuss this issue with their patients. This article objectively reviews the most common unconventional therapies used by cancer patients.
Collapse
|
28
|
Liu S, Cao X, Wang D, Zhu H. Iron metabolism: State of the art in hypoxic cancer cell biology. Arch Biochem Biophys 2022; 723:109199. [DOI: 10.1016/j.abb.2022.109199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 02/08/2023]
|
29
|
Mazrad ZAI, Schelle B, Nicolazzo JA, Leiske MN, Kempe K. Nitrile-Functionalized Poly(2-oxazoline)s as a Versatile Platform for the Development of Polymer Therapeutics. Biomacromolecules 2021; 22:4618-4632. [PMID: 34647734 DOI: 10.1021/acs.biomac.1c00923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In recent years, polymers bearing reactive groups have received significant interest for biomedical applications. Numerous functional polymer platforms have been introduced, which allow for the preparation of materials with tailored properties via post-polymerization modifications. However, because of their reactivity, many functional groups are not compatible with the initial polymerization. The nitrile group is a highly interesting and relatively inert functionality that has mainly received attention in radical polymerizations. In this Article, a nitrile-functionalized 2-oxazoline monomer (2-(4-nitrile-butyl)-2-oxazoline, BuNiOx) is introduced, and its compatibility with the cationic ring-opening polymerization is demonstrated. Subsequently, the versatility of nitrile-functionalized poly(2-oxazoline)s (POx) is presented. To this end, diverse (co)polymers are synthesized and characterized by nuclear resonance spectroscopy, size-exclusion chromatography, and mass spectrometry. Amphiphilic block copolymers are shown to efficiently encapsulate the hydrophobic drug curcumin (CUR) in aqueous solution, and the anti-inflammatory effect of the CUR-containing nanostructures is presented in BV-2 microglia. Furthermore, the availability of the BuNiOx repeating units for post-polymerization modifications with hydroxylamine to yield amidoxime (AO)-functionalized POx is demonstrated. These AO-containing POx were successfully applied for the complexation of Fe(III) in a quantitative manner. In addition, AO-functionalized POx were shown to release nitric oxide intracellularly in BV-2 microglia. Thus nitrile-functionalized POx represent a promising and robust platform for the design of polymer therapeutics for a wide range of applications.
Collapse
Affiliation(s)
- Zihnil A I Mazrad
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Baptiste Schelle
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Meike N Leiske
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
30
|
Guo H, Nomoto T, Muttaqien SE, Sun X, Komoto K, Matsui M, Miura Y, Nishiyama N. Polymeric Iron Chelators Enhancing Pro-Oxidant Antitumor Efficacy of Vitamin C by Inhibiting the Extracellular Fenton Reaction. Mol Pharm 2021; 18:4475-4485. [PMID: 34726400 DOI: 10.1021/acs.molpharmaceut.1c00673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Intravenously injected high-dose vitamin C (VC) induces extracellular H2O2, which can penetrate into the tumor cells and suppress tumor growth. However, extracellular labile iron ions in the tumor decompose H2O2 via the Fenton reaction, limiting the therapeutic effect. In this regard, we recently developed a polymeric iron chelator that can inactivate the intratumoral labile iron ions. Here, we examined the effect of our polymeric iron chelator on the high-dose VC therapy in in vitro and in vivo. In the in vitro study, the polymeric iron chelator could inactivate the extracellular labile iron ions and prevent the unfavorable decomposition of VC-induced H2O2, augmenting pro-oxidative damage to DNA and inducing apoptosis in cultured cancer cells. Even in the in vivo study, the polymeric iron chelator significantly improved the antitumor effect of VC in subcutaneous DLD-1 and CT26 tumors in mice, while conventional iron chelators could not. This work indicates the importance of modulating tumor-associated iron ions in the high-dose VC therapy and should contribute to a better understanding of its mechanism.
Collapse
Affiliation(s)
- Haochen Guo
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Sjaikhurrizal El Muttaqien
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Center for Pharmaceutical and Medical Technology, Agency for the Assessment and Application of Technology (BPPT), LAPTIAB I, PUSPITEK, Serpong, Banten 15314, Indonesia
| | - Xiaohang Sun
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Kana Komoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| |
Collapse
|
31
|
Chekmarev J, Azad MG, Richardson DR. The Oncogenic Signaling Disruptor, NDRG1: Molecular and Cellular Mechanisms of Activity. Cells 2021; 10:cells10092382. [PMID: 34572031 PMCID: PMC8465210 DOI: 10.3390/cells10092382] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
NDRG1 is an oncogenic signaling disruptor that plays a key role in multiple cancers, including aggressive pancreatic tumors. Recent studies have indicated a role for NDRG1 in the inhibition of multiple tyrosine kinases, including EGFR, c-Met, HER2 and HER3, etc. The mechanism of activity of NDRG1 remains unclear, but to impart some of its functions, NDRG1 binds directly to key effector molecules that play roles in tumor suppression, e.g., MIG6. More recent studies indicate that NDRG1s-inducing drugs, such as novel di-2-pyridylketone thiosemicarbazones, not only inhibit tumor growth and metastasis but also fibrous desmoplasia, which leads to chemotherapeutic resistance. The Casitas B-lineage lymphoma (c-Cbl) protein may be regulated by NDRG1, and is a crucial E3 ligase that regulates various protein tyrosine and receptor tyrosine kinases, primarily via ubiquitination. The c-Cbl protein can act as a tumor suppressor by promoting the degradation of receptor tyrosine kinases. In contrast, c-Cbl can also promote tumor development by acting as a docking protein to mediate the oncogenic c-Met/Crk/JNK and PI3K/AKT pathways. This review hypothesizes that NDRG1 could inhibit the oncogenic function of c-Cbl, which may be another mechanism of its tumor-suppressive effects.
Collapse
Affiliation(s)
- Jason Chekmarev
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (J.C.); (M.G.A.)
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (J.C.); (M.G.A.)
| | - Des R. Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (J.C.); (M.G.A.)
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Correspondence: ; Tel.: +61-7-3735-7549
| |
Collapse
|
32
|
Harismah K, Hajali N, Zandi H. 6-Thioguanine bimolecular formation for dual chelation of iron: DFT study. COMPUT THEOR CHEM 2021. [DOI: 10.1016/j.comptc.2021.113308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Mahajan SG, Nandre VS, Kodam KM, Kulkarni MV. Desferrioxamine E produced by an indigenous salt tolerant Pseudomonas stutzeri stimulates iron uptake of Triticum aestivum L. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2021. [DOI: 10.1016/j.bcab.2021.102057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
Marzo T, La Mendola D. The Effects on Angiogenesis of Relevant Inorganic Chemotherapeutics. Curr Top Med Chem 2021; 21:73-86. [PMID: 33243124 DOI: 10.2174/1568026620666201126163436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiogenesis is a key process allowing the formation of blood vessels. It is crucial for all the tissues and organs, ensuring their function and growth. Angiogenesis is finely controlled by several mechanisms involving complex interactions between pro- or antiangiogenic factors, and an imbalance in this control chain may result in pathological conditions. Metals as copper, zinc and iron cover an essential role in regulating angiogenesis, thus therapies having physiological metals as target have been proposed. In addition, some complexes of heavier metal ions (e.g., Pt, Au, Ru) are currently used as established or experimental anticancer agents targeting genomic or non-genomic targets. These molecules may affect the angiogenic mechanisms determining different effects that have been only poorly and non-systematically investigated so far. Accordingly, in this review article, we aim to recapitulate the impact on the angiogenic process of some reference anticancer drugs, and how it is connected to the overall pharmacological effects. In addition, we highlight how the activity of these drugs can be related to the role of biological essential metal ions. Overall, this may allow a deeper description and understanding of the antineoplastic activity of both approved or experimental metal complexes, providing important insights for the synthesis of new inorganic drugs able to overcome resistance and recurrence phenomena.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126, Pisa, Italy
| |
Collapse
|
35
|
Abbasi U, Abbina S, Gill A, Takuechi LE, Kizhakkedathu JN. Role of Iron in the Molecular Pathogenesis of Diseases and Therapeutic Opportunities. ACS Chem Biol 2021; 16:945-972. [PMID: 34102834 DOI: 10.1021/acschembio.1c00122] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Iron is an essential mineral that serves as a prosthetic group for a variety of proteins involved in vital cellular processes. The iron economy within humans is highly conserved in that there is no proper iron excretion pathway. Therefore, iron homeostasis is highly evolved to coordinate iron acquisition, storage, transport, and recycling efficiently. A disturbance in this state can result in excess iron burden in which an ensuing iron-mediated generation of reactive oxygen species imparts widespread oxidative damage to proteins, lipids, and DNA. On the contrary, problems in iron deficiency either due to genetic or nutritional causes can lead to a number of iron deficiency disorders. Iron chelation strategies have been in the works since the early 1900s, and they still remain the most viable therapeutic approach to mitigate the toxic side effects of excess iron. Intense investigations on improving the efficacy of chelation strategies while being well tolerated and accepted by patients have been a particular focus for many researchers over the past 30 years. Moreover, recent advances in our understanding on the role of iron in the pathogenesis of different diseases (both in iron overload and iron deficiency conditions) motivate the need to develop new therapeutics. We summarized recent investigations into the role of iron in health and disease conditions, iron chelation, and iron delivery strategies. Information regarding small molecule as well as macromolecular approaches and how they are employed within different disease pathogenesis such as primary and secondary iron overload diseases, cancer, diabetes, neurodegenerative diseases, infections, and in iron deficiency is provided.
Collapse
Affiliation(s)
- Usama Abbasi
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z7
| | - Srinivas Abbina
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z7
| | - Arshdeep Gill
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z1
| | - Lily E. Takuechi
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z7
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood Research, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z7
- Department of Chemistry, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z1
- The School of Biomedical Engineering, The University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| |
Collapse
|