1
|
Mrozińska Z, Kaczmarek A, Świerczyńska M, Juszczak M, Kudzin MH. Biochemical Behavior, Influence on Cell DNA Condition, and Microbiological Properties of Wool and Wool-Copper Materials. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2878. [PMID: 38930247 PMCID: PMC11204859 DOI: 10.3390/ma17122878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
The paper presents the study concerning the preparation and physio-chemical and biological properties of wool-copper (WO-Cu) materials obtained by the sputter deposition of copper onto the wool fibers. The WO-Cu material was subjected to physio-chemical and biological investigations. The physio-chemical investigations included the elemental analysis of materials (C, N, O, S, and Cu), their microscopic analysis, and surface properties analysis (specific surface area and total pore volume). The biological investigations consisted of the antimicrobial activity tests of the WO-Cu materials against colonies of Gram-positive (Staphylococcus aureus) bacteria, Gram-negative (Escherichia coli) bacteria, and fungal mold species (Chaetomium globosum). Biochemical-hematological tests included the evaluation of the activated partial thromboplastin time and pro-thrombin time. The tested wool-copper demonstrated the ability to interact with the DNA in a time-dependent manner. These interactions led to the DNA's breaking and degradation. The antimicrobial and antifungal activities of the WO-Cu materials suggest a potential application as an antibacterial/antifungal material. Wool-copper materials may be also used as customized materials where the blood coagulation process could be well controlled through the appropriate copper content.
Collapse
Affiliation(s)
- Zdzisława Mrozińska
- Łukasiewicz Research Network—Lodz Institute of Technology, 19/27 Marii Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (Z.M.); (M.J.)
| | - Anna Kaczmarek
- Łukasiewicz Research Network—Lodz Institute of Technology, 19/27 Marii Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (Z.M.); (M.J.)
| | - Małgorzata Świerczyńska
- Łukasiewicz Research Network—Lodz Institute of Technology, 19/27 Marii Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (Z.M.); (M.J.)
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, 90-537 Lodz, Poland
| | - Michał Juszczak
- Łukasiewicz Research Network—Lodz Institute of Technology, 19/27 Marii Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (Z.M.); (M.J.)
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Marcin H. Kudzin
- Łukasiewicz Research Network—Lodz Institute of Technology, 19/27 Marii Sklodowskiej-Curie Str., 90-570 Lodz, Poland; (Z.M.); (M.J.)
| |
Collapse
|
2
|
Yang L, Wang K, Guo L, Hu X, Zhou M. Unveiling the potential of HKUST-1: synthesis, activation, advantages and biomedical applications. J Mater Chem B 2024; 12:2670-2690. [PMID: 38411271 DOI: 10.1039/d3tb02929h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Metal-organic frameworks (MOFs) have emerged as a unique class of nanostructured materials, resulting from the self-assembly of metal ions or clusters with organic ligands, offering a wide range of applications in fields such as drug delivery, gas catalysis, and electrochemical sensing. Among them, HKUST-1, a copper-based MOF, has gained substantial attention due to its remarkable three-dimensional porous structure. Comprising copper ions and benzene-1,3,5-tricarboxylic acid, HKUST-1 exhibits an extraordinary specific surface area and pronounced porosity, making it a promising candidate in biomedicine. Notably, the incorporation of copper ions endows HKUST-1 with noteworthy activities, including antitumor, antibacterial, and wound healing-promoting properties. In this comprehensive review, we delve into the various synthesis methods and activation pathways employed in the preparation of HKUST-1. We also explore the distinct advantages of HKUST-1 in terms of its structural properties and functionalities. Furthermore, we investigate the exciting and rapidly evolving biomedical applications of HKUST-1. From its role in tumor treatment to its antibacterial effects and its ability to promote wound healing, we showcase the multifaceted potential of HKUST-1 in addressing critical challenges in biomedicine.
Collapse
Affiliation(s)
- Liuxuan Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ke Wang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Guo
- National Engineering Technology Research Center for Miao Medicine, Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiao Hu
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
3
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
4
|
Tsymbal S, Refeld A, Zatsepin V, Kuchur O. The p53 protein is a suppressor of Atox1 copper chaperon in tumor cells under genotoxic effects. PLoS One 2023; 18:e0295944. [PMID: 38127999 PMCID: PMC10735018 DOI: 10.1371/journal.pone.0295944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023] Open
Abstract
The p53 protein is crucial for regulating cell survival and apoptosis in response to DNA damage. However, its influence on therapy effectiveness is controversial: when DNA damage is high p53 directs cells toward apoptosis, while under moderate genotoxic stress it saves the cells from death and promote DNA repair. Furthermore, these processes are influenced by the metabolism of transition metals, particularly copper since they serve as cofactors for critical enzymes. The metallochaperone Atox1 is under intensive study in this context because it serves as transcription factor allegedly mediating described effects of copper. Investigating the interaction between p53 and Atox1 could provide insights into tumor cell survival and potential therapeutic applications in oncology. This study explores the relationship between p53 and Atox1 in HCT116 and A549 cell lines with wild type and knockout TP53. The study found an inverse correlation between Atox1 and p53 at the transcriptional and translational levels in response to genotoxic stress. Atox1 expression decreased with increased p53 activity, while cells with inactive p53 had significantly higher levels of Atox1. Suppression of both genes increased apoptosis, while suppression of the ATOX1 gene prevented apoptosis even under the treatment with chemotherapeutic drugs. The findings suggest that Atox1 may act as one of key elements in promotion of cell cycle under DNA-damaging conditions, while p53 works as an antagonist by inhibiting Atox1. Understanding of this relationship could help identify potential targets in cell signaling pathways to enhance the effectiveness of combined antitumor therapy, especially in tumors with mutant or inactive p53.
Collapse
Affiliation(s)
- Sergey Tsymbal
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, St. Petersburg, Russia
| | - Aleksandr Refeld
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, St. Petersburg, Russia
| | | | - Oleg Kuchur
- International Institute ‘Solution Chemistry of Advanced Materials and Technologies’, ITMO University, St. Petersburg, Russia
| |
Collapse
|
5
|
Zeng HL, Li H, Yang Q, Li CX. Transcriptomic Characterization of Copper-Binding Proteins for Predicting Prognosis in Glioma. Brain Sci 2023; 13:1460. [PMID: 37891828 PMCID: PMC10605646 DOI: 10.3390/brainsci13101460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/25/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Copper and copper-binding proteins are key components of tumor progression as they play important roles in tumor invasion and migration, but their associations in gliomas remain unclear. METHODS Transcriptomic datasets of glioblastoma, low-grade glioma, and normal brain cortex were derived from the TCGA and GTEX databases. Differentially expressed genes (DEGs) of copper-binding proteins were screened and used to construct a prognostic model based on COX and LASSO regression, which was further validated by the CGGA datasets. The expressions of risk-model genes were selectively confirmed via anatomic feature-based expression analysis and immunohistochemistry. The risk score was stratified by age, gender, WHO grade, IDH1 mutation, MGMT promoter methylation, and 1p/19q codeletion status, and a nomogram was constructed and validated. RESULTS A total of 21 DEGs of copper-binding proteins were identified and a six-gene risk-score model was constructed, consisting of ANG, F5, IL1A, LOXL1, LOXL2, and STEAP3, which accurately predicted 1-, 3-, and 5-year overall survival rates, with the AUC values of 0.87, 0.88, and 0.82, respectively. The high-risk group had a significantly shorter OS (p < 0.0001) and was associated with old age, wild-type IDH1, a high WHO grade, an unmethylated MGMT promoter, and 1p/19q non-codeletion and had higher levels of immune cell infiltration, cancer-immunity suppressor, and immune checkpoint gene expression as well as a higher TMB. CONCLUSIONS The model based on the genes of copper-binding proteins could contribute to prognosis prediction and provide potential targets against gliomas.
Collapse
Affiliation(s)
- Hao-Long Zeng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Huijun Li
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Qing Yang
- Institute of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China;
| | - Chao-Xi Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| |
Collapse
|
6
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
7
|
Lin Z, He Y, Wu Z, Yuan Y, Li X, Luo W. Comprehensive analysis of copper-metabolism-related genes about prognosis and immune microenvironment in osteosarcoma. Sci Rep 2023; 13:15059. [PMID: 37700003 PMCID: PMC10497601 DOI: 10.1038/s41598-023-42053-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Despite being significant in various diseases, including cancers, the impact of copper metabolism on osteosarcoma (OS) remains largely unexplored. This study aimed to use bioinformatics analyses to identify a reliable copper metabolism signature that could improve OS patient prognosis prediction, immune landscape understanding, and drug sensitivity. Through nonnegative matrix factorization (NMF) clustering, we revealed distinct prognosis-associated clusters of OS patients based on copper metabolism-related genes (CMRGs), showing differential gene expression linked to immune processes. The risk model, comprising 13 prognostic CMRGs, was established using least absolute shrinkage and selection operator (LASSO) Cox regression, closely associated with the OS microenvironment's immune situation and drug sensitivity. Furthermore, we developed an integrated nomogram, combining the risk score and clinical traits to quantitatively predict OS patient prognosis. The calibration plot, timeROC, and timeROC analyses demonstrated its predictable accuracy and clinical usefulness. Finally, we identified three independent prognostic signatures for OS patients: COX11, AP1B1, and ABCB6. This study confirmed the involvement of CMRGs in OS patient prognosis, immune processes, and drug sensitivity, suggesting their potential as promising prognostic signatures and therapeutic targets for OS.
Collapse
Affiliation(s)
- Zili Lin
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yizhe He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Ziyi Wu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Yuhao Yuan
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xiangyao Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
8
|
Yang S, Song Y, Hu Y, Chen H, Yang D, Song X. Multifaceted Roles of Copper Ions in Anticancer Nanomedicine. Adv Healthc Mater 2023; 12:e2300410. [PMID: 37027332 DOI: 10.1002/adhm.202300410] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/27/2023] [Indexed: 04/08/2023]
Abstract
The significantly increased copper level in tumor tissues and serum indicates the close association of copper ions with tumor development, making copper ions attractive targets in the development of novel tumor treatment methods. The advanced nanotechnology developed in the past decades provides great potential for tumor therapy, among which Cu-based nanotherapeutic systems have received greater attention. Herein, the multifaceted roles of copper ions in cancer progression are summarized and the recent advances in the copper-based nanostructures or nanomedicines for different kinds of tumor therapies including copper depletion therapy, copper-based cytotoxins, copper-ion-based chemodynamic therapy and its combination with other treatments, and copper-ion-induced ferroptosis and cuproptosis activation are discussed. Furthermore, the perspectives for the further development of copper-ion-based nanomedicines for tumor therapy and clinic translation are presented by the authors.
Collapse
Affiliation(s)
- Siyuan Yang
- Department of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, P. R. China
| | - Yingnan Song
- Department of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, P. R. China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, 550025, P. R. China
| | - Yanling Hu
- Nanjing Polytechnic Institute, 210048, Nanjing, China
| | - HongJin Chen
- Department of Cardiac Surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550009, P. R. China
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, 550025, P. R. China
| | - Dongliang Yang
- School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), 211816, 30 South Puzhu Road, Nanjing, China
| | - Xuejiao Song
- School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), 211816, 30 South Puzhu Road, Nanjing, China
| |
Collapse
|
9
|
Qi B, Xu Q, Cao Y, Xiao Z. Photothermal and Catalytic Performance of Multifunctional Cu-Fe Bimetallic Prussian Blue Nanocubes with the Assistance of Near-Infrared Radiation. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1897. [PMID: 37446413 DOI: 10.3390/nano13131897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023]
Abstract
Copper and iron are the basic metal elements that have attracted much attention in industry. Prussian blue (PB) is a significant class of metal-organic frameworks (MOFs); however, the lack of such linkages between the structure and properties, as well as properties differences, limits their potential applications. In this paper, the Cu-based Prussian blue nanocubes with and without Fe doping were synthesized. With the increasing reaction time, the morphology of the Cu-based Prussian blue nanocubes without Fe doping (PB:Cu NCs) changes from cuboidal to circular, and finally grows back to cuboidal. However, Cu-based Prussian blue nanocubes with Fe doping (PB: CuFe NCs) grow directly from the cube and eventually collapse. The nanocubes show a notable red shift with the tunable spectra from 400 nm to 700 nm. Compared with PB: Cu NCs, the PB: CuFe NCs have higher temperature rise under 808 nm irradiation and better photothermal efficacy. The catalytic efficiency of PB: CuFe NCs changes with the pH and reaches its maximum value of 1.021 mM with a pH of 5.5. The enhanced catalytic reaction by the near-infrared radiation plasmonic photothermal effect is also confirmed. This work highlights the potential of the developed PB: Cu and PB: CuFe NCs for photothermal-enhanced co-catalysis nanomaterials.
Collapse
Affiliation(s)
- Bairui Qi
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Qiang Xu
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Yunxuan Cao
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Zhu Xiao
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
- Key Laboratory of Non-Ferrous Metal Materials Science and Engineering, Ministry of Education, Changsha 410083, China
| |
Collapse
|
10
|
Tsymbal SA, Refeld AG, Kuchur OA. The p53 Tumor Suppressor and Copper Metabolism: An Unrevealed but Important Link. Mol Biol 2022. [DOI: 10.1134/s0026893322060188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
11
|
Cun JE, Fan X, Pan Q, Gao W, Luo K, He B, Pu Y. Copper-based metal-organic frameworks for biomedical applications. Adv Colloid Interface Sci 2022; 305:102686. [PMID: 35523098 DOI: 10.1016/j.cis.2022.102686] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Metal-organic frameworks (MOFs) are a class of important porous, crystalline materials composed of metal ions (clusters) and organic ligands. Owing to the unique redox chemistry, photochemical and electrical property, and catalytic activity of Cu2+/+, copper-based MOFs (Cu-MOFs) have been recently and extensively explored in various biomedical fields. In this review, we first make a brief introduction to the synthesis of Cu-MOFs and their composites, and highlight the recent synthetic strategies of two most studied representatives, three-dimensional HKUST-1 and two-dimensional Cu-TCPP. The recent advances of Cu-MOFs in the applications of cancer treatment, bacterial inhibition, biosensing, biocatalysis, and wound healing are summarized and discussed. Furthermore, we propose a prospect of the future development of Cu-MOFs in biomedical fields and beyond.
Collapse
Affiliation(s)
- Ju-E Cun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
12
|
Mirhaj M, Labbaf S, Tavakoli M, Seifalian A. An Overview on the Recent Advances in the Treatment of Infected Wounds: Antibacterial Wound Dressings. Macromol Biosci 2022; 22:e2200014. [PMID: 35421269 DOI: 10.1002/mabi.202200014] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/20/2022] [Indexed: 11/11/2022]
Abstract
A wound can be surgical, cuts from an operation or due to accident and trauma. The infected wound, as a result of bacteria growth within the damaged skin, interrupts the natural wound healing process and significantly impacts the quality of life. Wound dressing is an important segment of the skincare industry with its economic burden estimated at $ 20.4 billion (in 2021) in the global market. The results of recent clinical trials suggest that the use of modern dressings can be the easiest, most accessible, and most cost-effective way to treat chronic wounds and, hence, holds significant promise. With the sheer number of dressings in the market, the selection of correct dressing is confusing for clinicians and healthcare workers. The aim of this research was to review widely used types of antibacterial wound dressings, as well as emerging products, for their efficiency and mode of action. In this review, we focus on introducing antibiotics and antibacterial nanoparticles as two important and clinically widely used categories of antibacterial agents. The perspectives and challenges for paving the way for future research in this field are also discussed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marjan Mirhaj
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Mohamadreza Tavakoli
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Amelia Seifalian
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Del Bello F, Pellei M, Bagnarelli L, Santini C, Giorgioni G, Piergentili A, Quaglia W, Battocchio C, Iucci G, Schiesaro I, Meneghini C, Venditti I, Ramanan N, De Franco M, Sgarbossa P, Marzano C, Gandin V. Cu(I) and Cu(II) Complexes Based on Lonidamine-Conjugated Ligands Designed to Promote Synergistic Antitumor Effects. Inorg Chem 2022; 61:4919-4937. [PMID: 35285628 PMCID: PMC8965879 DOI: 10.1021/acs.inorgchem.1c03658] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bis(pyrazol-1-yl)- and bis(3,5-dimethylpyrazol-1-yl)-acetates were conjugated with the 2-hydroxyethylester and 2-aminoethylamide derivatives of the antineoplastic drug lonidamine to prepare Cu(I) and Cu(II) complexes that might act through synergistic mechanisms of action due to the presence of lonidamine and copper in the same chemical entity. Synchrotron radiation-based complementary techniques [X-ray photorlectron spectroscopy and near-edge X-ray absorption fine structure (NEXAFS)] were used to characterize the electronic and molecular structures of the complexes and the local structure around the copper ion (XAFS) in selected complexes. All complexes showed significant antitumor activity, proving to be more effective than the reference drug cisplatin in a panel of human tumor cell lines, and were able to overcome oxaliplatin and multidrug resistance. Noticeably, these Cu complexes appeared much more effective than cisplatin against 3D spheroids of pancreatic PSN-1 cancer cells; among these, PPh3-containing Cu(I) complex 15 appeared to be the most promising derivative. Mechanistic studies revealed that 15 induced cancer cell death by means of an apoptosis-alternative cell death.
Collapse
Affiliation(s)
- Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Maura Pellei
- School of Science and Technology, Chemistry Division, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Luca Bagnarelli
- School of Science and Technology, Chemistry Division, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Carlo Santini
- School of Science and Technology, Chemistry Division, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino 1, 62032 Camerino, Italy
| | - Chiara Battocchio
- Department of Science, Roma Tre University, Via della Vasca Navale 79, 00146 Roma, Italy
| | - Giovanna Iucci
- Department of Science, Roma Tre University, Via della Vasca Navale 79, 00146 Roma, Italy
| | - Irene Schiesaro
- Department of Science, Roma Tre University, Via della Vasca Navale 79, 00146 Roma, Italy
| | - Carlo Meneghini
- Department of Science, Roma Tre University, Via della Vasca Navale 79, 00146 Roma, Italy
| | - Iole Venditti
- Department of Science, Roma Tre University, Via della Vasca Navale 79, 00146 Roma, Italy
| | - Nitya Ramanan
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, U.K
| | - Michele De Franco
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Paolo Sgarbossa
- Department of Industrial Engineering, University of Padova, via Marzolo 9, 35131 Padova, Italy
| | - Cristina Marzano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
14
|
Oliveri V. Selective Targeting of Cancer Cells by Copper Ionophores: An Overview. Front Mol Biosci 2022; 9:841814. [PMID: 35309510 PMCID: PMC8931543 DOI: 10.3389/fmolb.2022.841814] [Citation(s) in RCA: 197] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 12/15/2022] Open
Abstract
Conventional cancer therapies suffer from severe off-target effects because most of them target critical facets of cells that are generally shared by all rapidly proliferating cells. The development of new therapeutic agents should aim to increase selectivity and therefore reduce side effects. In addition, these agents should overcome cancer cell resistance and target cancer stem cells. Some copper ionophores have shown promise in this direction thanks to an intrinsic selectivity in preferentially inducing cuproptosis of cancer cells compared to normal cells. Here, Cu ionophores are discussed with a focus on selectivity towards cancer cells and on the mechanisms responsible for this selectivity. The proposed strategies, to further improve the targeting of cancer cells by copper ionophores, are also reported.
Collapse
|
15
|
Liu J, Yuan Y, Cheng Y, Fu D, Chen Z, Wang Y, Zhang L, Yao C, Shi L, Li M, Zhou C, Zou M, Wang G, Wang L, Wang Z. Copper-Based Metal-Organic Framework Overcomes Cancer Chemoresistance through Systemically Disrupting Dynamically Balanced Cellular Redox Homeostasis. J Am Chem Soc 2022; 144:4799-4809. [PMID: 35192770 DOI: 10.1021/jacs.1c11856] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemodrug resistance is a major reason accounting for tumor recurrence. Given the mechanistic complexity of chemodrug resistance, molecular inhibitors and targeting drugs often fail to eliminate drug-resistant cancer cells, and sometimes even promote chemoresistance by activating alternative pathways. Here, by exploiting biochemical fragility of high-level but dynamically balanced cellular redox homeostasis in drug-resistant cancer cells, we design a nanosized copper/catechol-based metal-organic framework (CuHPT) that effectively disturbs this homeostasis tilting the balance toward oxidative stress. Within drug-resistant cells, CuHPT starts disassembly that is triggered by persistent consumption of cellular glutathione (GSH). CuHPT disassembly simultaneously releases two structural elements: catechol ligands and reductive copper ions (Cu+). Both of them cooperatively function to amplify the production of intracellular radical oxidative species (ROS) via auto-oxidation and Fenton-like reactions through exhausting GSH. By drastically heightening cellular oxidative stress, CuHPT exhibits selective and potent cytotoxicity to multiple drug-resistant cancer cells. Importantly, CuHPT effectively inhibits in vivo drug-resistant tumor growth and doubles the survival time of tumor-bearing mice. Thus, along with CuHPT's good biocompatibility, our biochemical, cell biological, preclinical animal model data provide compelling evidence supporting the notion that this copper-based MOF is a predesigned smart therapeutic against drug-resistant cancers through precisely deconstructing their redox homeostasis.
Collapse
Affiliation(s)
- Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ye Yuan
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanni Cheng
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Daan Fu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhongyin Chen
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lifang Zhang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chundong Yao
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Shi
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mingyi Li
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cheng Zhou
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Meizhen Zou
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
16
|
Yang Z, Deng W, Zhang X, An Y, Liu Y, Yao H, Zhang Z. Opportunities and Challenges of Nanoparticles in Digestive Tumours as Anti-Angiogenic Therapies. Front Oncol 2022; 11:789330. [PMID: 35083147 PMCID: PMC8784389 DOI: 10.3389/fonc.2021.789330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/10/2021] [Indexed: 01/04/2023] Open
Abstract
Digestive tumours, a common kind of malignancy worldwide, have recently led to the most tumour-related deaths. Angiogenesis, the process of forming novel blood vessels from pre-existing vessels, is involved in various physiological and pathological processes in the body. Many studies suggest that abnormal angiogenesis plays an important role in the growth, progression, and metastasis of digestive tumours. Therefore, anti-angiogenic therapy is considered a promising target for improving therapeutic efficacy. Traditional strategies such as bevacizumab and regorafenib can target and block the activity of proangiogenic factors to treat digestive tumours. However, due to resistance and some limitations, such as poor pharmacokinetics, their efficacy is not always satisfactory. In recent years, nanotechnology-based anti-angiogenic therapies have emerged as a new way to treat digestive tumours. Compared with commonly used drugs, nanoparticles show great potential in tumour targeted delivery, controlled drug release, prolonged cycle time, and increased drug bioavailability. Therefore, anti-angiogenic nanoparticles may be an effective complementary therapy to treat digestive tumours. In this review, we outline the different mechanisms of angiogenesis, the effects of nanoparticles on angiogenesis, and their biomedical applications in various kinds of digestive tumours. In addition, the opportunities and challenges are briefly discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
17
|
Lee D, Ha J, Kang M, Yang Z, Jiang W, Kim BYS. Strategies of Perturbing Ion Homeostasis for Cancer Therapy. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- DaeYong Lee
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - JongHoon Ha
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Minjeong Kang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Zhaogang Yang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Wen Jiang
- Department of Radiation Oncology The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| | - Betty Y. S. Kim
- Department of Neurosurgery The University of Texas MD Anderson Cancer Center Houston TX 77030 USA
| |
Collapse
|
18
|
Geng Y, Sun R, Zhang Y, Zhou Z, Shen Y. Copper (Ⅱ)/cis-platinum -loaded nanogels as an adjuvant potentiate disulfiram antitumor efficacy. Biomater Sci 2022; 10:1384-1392. [DOI: 10.1039/d1bm01795k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Disulfiram (DSF) is nontoxic and exerts anticancer activity by forming highly toxic chelate via its metabolite diethyldithiocarbamate with transition metal ions. However, there are not enough such ions in the...
Collapse
|
19
|
Leveraging disulfiram to treat cancer: Mechanisms of action, delivery strategies, and treatment regimens. Biomaterials 2021; 281:121335. [PMID: 34979419 DOI: 10.1016/j.biomaterials.2021.121335] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/07/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023]
Abstract
Disulfiram (DSF) has been used as an alcoholism drug for 70 years. Recently, it has attracted increasing attention owing to the distinguished anticancer activity, which can be further potentiated by the supplementation of Cu2+. Although encouraging anticancer results are obtained in lab, the clinical outcomes of oral DSF are not satisfactory, which urges an in-depth understanding of the underlying mechanisms, bottlenecks, and proposal of potential methods to address the dilemma. In this review, a critical summarization of various molecular biological anticancer mechanisms of DSF/Cu2+ is provided and the predicament of orally delivering DSF in clinical oncotherapy is explained by the metabolic barriers. We highlight the recent advances in the DSF/Cu2+ delivery strategies and the emerging treatment regimens for cancer treatment. Last but not the least, we summarize the clinical trials regarding DSF and make a prospect of DSF/Cu-based cancer therapy.
Collapse
|
20
|
Chen M, Huang Z, Xia M, Ding Y, Shan T, Guan Z, Dai X, Xu X, Huang Y, Huang M, Zhao C. Glutathione-responsive copper-disulfiram nanoparticles for enhanced tumor chemotherapy. J Control Release 2021; 341:351-363. [PMID: 34856225 DOI: 10.1016/j.jconrel.2021.11.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022]
Abstract
Disulfiram (DSF), a familiar FDA-approved drug used for alcohol withdrawal, has recently been verified with potent antitumor therapeutic effect by generating Cu(DTC)2, which is the complex of its metabolite diethyldithiocarbamate (DTC) and copper. However, its poor tumor selectivity and insufficient endogenous Cu2+ concentration within tumor site largely hinders the application of DSF-based antitumor therapy. Therefore, a GSH-responsive coordination nanoparticles (Cu-IXZ@DSF) was established as a copper carrier to achieve synchronous but separate delivery of Cu2+ and DSF without antitumor ability, further to realize selectively triggered tumor in situ Cu(DTC)2 generation for antitumor therapy. A widely-used proteasome inhibitor ixazomib (IXZ) was chosen as ligands and Cu2+ was used as coordination nodes to form nanosized Cu-IXZ@DSF. The DSF encapsulated in Cu-IXZ@DSF could be reduced to DTC by intracellular GSH, which could contend for Cu2+ and realize in situ high toxic Cu(DTC)2 generation. Meanwhile, the chelation could lead to the disassembly of Cu-IXZ@DSF and release of IXZ to eventually achieve tumor specific "transformation from low toxicity to high toxicity" chemotherapy. The results of in vitro and in vivo experiments demonstrated that the as-prepared nanoplatform Cu-IXZ@DSF showed good biosafety and excellent antitumor effect via endoplasmic reticulum stress (ERS) as well as reactive oxygen species (ROS) generation pathway. Therefore, this nanocarrier provides an inspiring strategy with specific-triggered antitumor Cu(DTC)2 generation for DSF-based chemotherapy with high therapeutic effect and biosafety and showing great potential of treating cancer.
Collapse
Affiliation(s)
- Meixu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Zeqian Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Meng Xia
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Yaqing Ding
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Ting Shan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Zilin Guan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Xiuling Dai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Xiaoyu Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
21
|
Massoud SS, Louka FR, Dial MT, Malek AJ, Fischer RC, Mautner FA, Vančo J, Malina T, Dvořák Z, Trávníček Z. Identification of potent anticancer copper(ii) complexes containing tripodal bis[2-ethyl-di(3,5-dialkyl-1H-pyrazol-1-yl)]amine moiety. Dalton Trans 2021; 50:11521-11534. [PMID: 34346447 DOI: 10.1039/d1dt01724a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of heteroleptic copper(ii) complexes of the composition [Cu(L1-5)Cl]X, where X = ClO4 and/or PF6 and [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl))-(6-methyl-(2-pyridylmethyl))]amine (L1), [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl))-(3,4-dimethoxy-(2-pyridylmethyl))]amine (L2), [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl)-(2-quinolymethyl)]amine (L3), [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazolyl)-(di(3,5-dimethyl-1H-pyrazol-1-yl-methyl))]amine (L4) and [bis(2-ethyl-di(3,5-dimethyl-1H-pyrazol-1-yl)-(5-methyl-3-phenyl-1H-pyrazol-1-yl-methyl)]amine (L5), were prepared and thoroughly characterized including single-crystal X-ray diffraction technique. The in vitro cytotoxicity of complexes against A2780, A2780R, HOS and MCF-7 human cancer cell lines was evaluated using the MTT test. The results revealed that complexes [Cu(L1)Cl]PF6 (1-PF6), [Cu(L2)Cl]ClO4 (2-ClO4) and [Cu(L3)Cl]PF6 (3-PF6) are the most effective, with IC50 values ranging from 1.4 to 6.3 μM, thus exceeding the cytotoxic potential of metallodrug cisplatin (IC50 values ranging from 29.9 to 82.0 μM). The complexes [Cu(L4)Cl]PF6 (4-PF6) and [Cu(L5)Cl]PF6 (5-PF6) showed only moderate cytotoxicity against A2780, with IC50 = 53.6 μM, and 33.8 μM, respectively. The cell cycle profile, time-resolved cellular uptake, interactions with small sulfur-containing biomolecules (cysteine and glutathione), intracellular ROS production, induction of apoptosis and activation of caspases 3/7 were also evaluated in the case of the selected complexes. It has been found that the best performing complexes 1 and 2 cause cell arrest in the G2/M phase and induce apoptosis via the increase in production of ROS, dominantly due to the overproduction of superoxide.
Collapse
Affiliation(s)
- Salah S Massoud
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 43700, Lafayette, LA 70504, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ji X, Zhou Y, Li Q, Song H, Fan C. Protein-Mimicking Nanoparticles for a Cellular Regulation of Homeostasis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:31331-31336. [PMID: 34227383 DOI: 10.1021/acsami.1c09281] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The distinct physical and chemical properties of nanoparticles (NPs) offer great opportunities to develop new strategies for diagnostic and therapeutic purposes. Whereas NPs often serve as inert nanocarriers, their inherent "biological" activities have recently been extensively unveiled and explored. These protein-mimicking NPs (dubbed protmins) have been reported to modulate a cellular homeostasis without displaying a general toxicity, which may act as potential nanomedicines to provide a monotherapy or combination therapy in a disease treatment. In the meanwhile, the unexpected behaviors of protmins in complex biological systems also raise new concerns on the biosafety issue. Herein, we summarize several categories of the protmin-based regulation of cellular homeostasis and discuss their broad effects on cell functions and behaviors.
Collapse
Affiliation(s)
- Xiaoyuan Ji
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanfeng Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
23
|
Liang P, Ballou B, Lv X, Si W, Bruchez MP, Huang W, Dong X. Monotherapy and Combination Therapy Using Anti-Angiogenic Nanoagents to Fight Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005155. [PMID: 33684242 DOI: 10.1002/adma.202005155] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/10/2020] [Indexed: 06/12/2023]
Abstract
Anti-angiogenic therapy, targeting vascular endothelial cells (ECs) to prevent tumor growth, has been attracting increasing attention in recent years, beginning with bevacizumab (Avastin) through its Phase II/III clinical trials on solid tumors. However, these trials showed only modest clinical efficiency; moreover, anti-angiogenic therapy may induce acquired resistance to the drugs employed. Combining advanced drug delivery techniques (e.g., nanotechnology) or other therapeutic strategies (e.g., chemotherapy, radiotherapy, phototherapy, and immunotherapy) with anti-angiogenic therapy results in significantly synergistic effects and has opened a new horizon in fighting cancer. Herein, clinical difficulties in using traditional anti-angiogenic therapy are discussed. Then, several promising applications of anti-angiogenic nanoagents in monotherapies and combination therapies are highlighted. Finally, the challenges and perspectives of anti-angiogenic cancer therapy are summarized. A useful introduction to anti-angiogenic strategies, which may significantly improve therapeutic outcomes, is thus provided.
Collapse
Affiliation(s)
- Pingping Liang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Byron Ballou
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA, 15213, United States
| | - Xinyi Lv
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Weili Si
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
| | - Marcel P Bruchez
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA, 15213, United States
| | - Wei Huang
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, China
- School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| |
Collapse
|
24
|
Gaál A, Garay TM, Horváth I, Máthé D, Szöllősi D, Veres DS, Mbuotidem J, Kovács T, Tóvári J, Bergmann R, Streli C, Szakács G, Mihály J, Varga Z, Szoboszlai N. Development and In Vivo Application of a Water-Soluble Anticancer Copper Ionophore System Using a Temperature-Sensitive Liposome Formulation. Pharmaceutics 2020; 12:pharmaceutics12050466. [PMID: 32443790 PMCID: PMC7284829 DOI: 10.3390/pharmaceutics12050466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Liposomes containing copper and the copper ionophore neocuproine were prepared and characterized for in vitro and in vivo anticancer activity. Thermosensitive PEGylated liposomes were prepared with different molar ratios of 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine (DPPC) and hydrogenated soybean phosphatidylcholine (HSPC) in the presence of copper(II) ions. Optimal, temperature dependent drug release was obtained at 70:30 DPPC to HSPC weight ratio. Neocuproine (applied at 0.2 mol to 1 mol phospholipid) was encapsulated through a pH gradient while using unbuffered solution at pH 4.5 inside the liposomes, and 100 mM HEPES buffer pH 7.8 outside the liposomes. Copper ions were present in excess, yielding 0.5 mM copper-(neocuproine)2 complex and 0.5 mM free copper. Pre-heating to 45 °C increased the toxicity of the heat-sensitive liposomes in short-term in vitro experiments, whereas at 72 h all investigated liposomes exhibited similar in vitro toxicity to the copper(II)-neocuproine complex (1:1 ratio). Thermosensitive liposomes were found to be more effective in reducing tumor growth in BALB/c mice engrafted with C26 cancer cells, regardless of the mild hyperthermic treatment. Copper uptake of the tumor was verified by PET/CT imaging following treatment with [64Cu]Cu-neocuproine liposomes. Taken together, our results demonstrate the feasibility of targeting a copper nanotoxin that was encapsulated in thermosensitive liposomes containing an excess of copper.
Collapse
Affiliation(s)
- Anikó Gaál
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; (A.G.); (J.M.)
| | - Tamás M. Garay
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, H-1083 Budapest, Práter utca 50/a, Hungary
- 1st Department of Internal Medicine and Oncology, Semmelweis University, H-1083 Budapest, Hungary
- Correspondence: (T.M.G.); (Z.V.); (N.S.); Tel.: +36-1-8864-769 (T.M.G.); +36-1-382-6568 (Z.V.); +36-1-372-2500 (ext. 6430) (N.S.)
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (I.H.); (D.M.); (D.S.); (D.S.V.); (R.B.)
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (I.H.); (D.M.); (D.S.); (D.S.V.); (R.B.)
- CROmed Translational Research Centers Ltd., H-1047 Budapest, Hungary
| | - Dávid Szöllősi
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (I.H.); (D.M.); (D.S.); (D.S.V.); (R.B.)
| | - Dániel S. Veres
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (I.H.); (D.M.); (D.S.); (D.S.V.); (R.B.)
| | - Jeremiah Mbuotidem
- Institute of Translational Medicine, Semmelweis University, H-1094 Budapest, Hungary;
| | - Tibor Kovács
- Institute of Radiochemistry and Radioecology, University of Pannonia, H-8200 Veszprém, Hungary;
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary;
| | - Ralf Bergmann
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary; (I.H.); (D.M.); (D.S.); (D.S.V.); (R.B.)
- Helmholz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, D-01328 Dresden, Germany
| | - Christina Streli
- Institute of Atomic and Subatomic Physics, Atominstitut, TU Wien, A-1020 Vienna, Stadionallee 2, Austria;
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary;
- Institute of Cancer Research, Medical University Vienna, A-1090 Vienna, Austria
| | - Judith Mihály
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; (A.G.); (J.M.)
| | - Zoltán Varga
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117 Budapest, Hungary; (A.G.); (J.M.)
- Correspondence: (T.M.G.); (Z.V.); (N.S.); Tel.: +36-1-8864-769 (T.M.G.); +36-1-382-6568 (Z.V.); +36-1-372-2500 (ext. 6430) (N.S.)
| | - Norbert Szoboszlai
- Laboratory for Environmental Chemistry and Bioanalytics, Institute of Chemistry, Eötvös Loránd University, H-1117 Budapest, Pázmány Péter Stny. 1/A, Hungary
- Correspondence: (T.M.G.); (Z.V.); (N.S.); Tel.: +36-1-8864-769 (T.M.G.); +36-1-382-6568 (Z.V.); +36-1-372-2500 (ext. 6430) (N.S.)
| |
Collapse
|
25
|
Serra M, Columbano A, Ammarah U, Mazzone M, Menga A. Understanding Metal Dynamics Between Cancer Cells and Macrophages: Competition or Synergism? Front Oncol 2020; 10:646. [PMID: 32426284 PMCID: PMC7203474 DOI: 10.3389/fonc.2020.00646] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
Metal ions, such as selenium, copper, zinc, and iron are naturally present in the environment (air, drinking water, and food) and are vital for cellular functions at chemical, molecular, and biological levels. These trace elements are involved in various biochemical reactions by acting as cofactors for many enzymes and control important biological processes by binding to the receptors and transcription factors. Moreover, they are essential for the stabilization of the cellular structures and for the maintenance of genome stability. A body of preclinical and clinical evidence indicates that dysregulation of metal homeostasis, both at intracellular and tissue level, contributes to the pathogenesis of many different types of cancer. These trace minerals play a crucial role in preventing or accelerating neoplastic cell transformation and in modulating the inflammatory and pro-tumorigenic response in immune cells, such as macrophages, by controlling a plethora of metabolic reactions. In this context, macrophages and cancer cells interact in different manners and some of these interactions are modulated by availability of metals. The current review discusses the new findings and focuses on the involvement of these micronutrients in metabolic and cellular signaling mechanisms that influence macrophage functions, onset of cancer and its progression. An improved understanding of "metallic" cross-talk between macrophages and cancer cells may pave the way for innovative pharmaceutical or dietary interventions in order to restore the balance of these trace elements and also strengthen the chemotherapeutic treatment.
Collapse
Affiliation(s)
- Marina Serra
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Amedeo Columbano
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| | - Alessio Menga
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| |
Collapse
|
26
|
Zhong Y, Sun R, Geng Y, Zhou Q, Piao Y, Xie T, Zhou R, Shen Y. N-Oxide polymer-cupric ion nanogels potentiate disulfiram for cancer therapy. Biomater Sci 2020; 8:1726-1733. [PMID: 31995039 DOI: 10.1039/c9bm01841g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Disulfiram (DSF) exerts potent anticancer activity via the formation of chelates with copper or zinc ions in tumor tissues, but the low abundance of these ions in the tumor cannot sustain its antitumor activity. Herein, we show that a zwitterionic water-soluble N-oxide polymer, poly[2-(N-oxide-N,N-dimethylamino)ethyl methacrylate] (OPDMA), can complex cupric ions and form nanogels (OPDMA/Cu), which efficiently deliver copper ions to tumor tissue to potentiate DSF significantly for effective antitumor therapy.
Collapse
Affiliation(s)
- Yin Zhong
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| | - Rui Sun
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| | - Yu Geng
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| | - Quan Zhou
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| | - Ying Piao
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| | - Tao Xie
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| | - Ruhong Zhou
- Institute of Quantitative Biology and Department of Physics, Zhejiang University, Hangzhou, 310027, China.
| | - Youqing Shen
- College of Chemical and Biological Engineering, Hangzhou, 310027, China.
| |
Collapse
|