1
|
Mohammadi Zonouz A, Taghavi S, Nekooei S, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. Synthesis of targeted doxorubicin-loaded gold nanorod -mesoporous manganese dioxide core-shell nanostructure for ferroptosis, chemo-photothermal therapy in vitro and in vivo. Int J Pharm 2024; 665:124725. [PMID: 39293581 DOI: 10.1016/j.ijpharm.2024.124725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/14/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
In the current study, a core-shell inorganic nanostructure comprising a gold nanorod core and -mesoporous manganese dioxide shell was synthesized. Then, the mesoporous manganese dioxide shell was loaded with doxorubicin (DOX) and then coated with pluronic F127 and pluronic F127-folic acid conjugate (1.5:1 wt ratio of pluronic F127: pluronic F127-folic acid conjugate) to prepare targeted final platform. In this design, mesoporous manganese dioxide acted as a reservoir for DOX loading, anti-hypoxia, and MRI contrast agent, while the gold nanorod core acted as a photothermal and CT scan imaging agent. DOX was encapsulated in the mesoporous manganese dioxide shell with a loading capacity and loading efficiency of 19.8 % ± 0.2 and 99.0 % ± 0.9, respectively. The in vitro release experiment showed the impact of glutathione (GSH), mildly acidic pH, and laser irradiating toward accelerated stimuli-responsive DOX release. The ·OH production of the prepared platform was verified by methylene blue (MB) decomposition reaction. Furthermore, thermal imaging exhibited the ability of the prepared platform to convert the NIR irradiation to heat. In vitro cytotoxicity tests on the folate receptor-positive 4 T1 cell line revealed the remarkable cytotoxicity of the targeted formulation compared to the nontargeted formulation (statistically significant). The MTT experiment demonstrated that exposure to laser 808 irradiation enhanced cytotoxicity of the targeted formulation (p < 0.0001). The production of ROS in 4 T1 cells following treatment with the targeted formulation was demonstrated by the dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay. Furthermore, in vivo investigations by implementing subcutaneous 4 T1 tumorized female BABL/c mice indicated that the prepared platform was an effective system in suppressing tumor growth by combining chemotherapy with PTT (photothermal therapy). Additionally, simultanous PTT and anti-hypoxic activity of this system showed potent tumor growth suppression impact. The percent of tumor size reduction in mice treated with FA-F127-DOX@Au-MnO2 + 808 nm laser compared to the control group was 99.7 %. The results of the biodistribution investigation showed tumor accumulation and modified pharmacokinetics of the targeted system. Lastly, 6 and 24 h post-intravenous injection, CT-scan and MR imagings capability of the prepared platform was verified in preclinical stage. The prepared multipurpose system introduces great opportunity to provide multiple treatment strategy along with multimodal imaging capability in a single platform for breast cancer treatment.
Collapse
Affiliation(s)
- Aidin Mohammadi Zonouz
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Taghavi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medicinal Chemistry Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Hu W, Zhao Z, Du J, Jiang J, Yang M, Tian M, Zhao P. Interferon signaling and ferroptosis in tumor immunology and therapy. NPJ Precis Oncol 2024; 8:177. [PMID: 39127858 DOI: 10.1038/s41698-024-00668-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
This study sought to elucidate the mechanisms underlying the impact of the interferon signaling pathway on Ferroptosis in tumor cells and its correlation with CD8 + T cell exhaustion. Using mouse models and single-cell sequencing, the researchers studied the interaction between CD8 + T cells and the interferon signaling pathway. Differential gene analysis revealed key genes involved in CD8 + T cell exhaustion, and their downstream factors were explored using bioinformatics tools. The expression levels of interferon-related genes associated with Ferroptosis were analyzed using data from the TCGA database, and their relevance to tumor tissue Ferroptosis and patients' prognosis was determined. In vitro experiments were conducted to measure the levels of IFN-γ, MDA, and LPO, as well as tumor cell viability and apoptosis. In vivo validation using a mouse tumor model confirmed the results obtained from the in vitro experiments, highlighting the potential of silencing HSPA6 or DNAJB1 in enhancing the efficacy of PD-1 therapy and inhibiting tumor growth and migration.
Collapse
Affiliation(s)
- Wei Hu
- Department of Breast Surgery, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, PR China
| | - Ziqian Zhao
- The Second Medical College, Xinjiang Medical University, Urumqii, PR China
| | - Jianxin Du
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, PR China
| | - Jie Jiang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, PR China
| | - Minghao Yang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, PR China
| | - Maojin Tian
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, PR China.
| | - Peiqing Zhao
- Center of Translational Medicine, Zibo Central Hospital Affiliated to Binzhou Medical University, Zibo, PR China.
| |
Collapse
|
3
|
Bernkop-Schnürch A, Chavooshi D, Descher HA, Leitner D, Talasz H, Hermann M, Wurst K, Hohloch S, Gust R, Kircher B. Design, Synthesis, Electrochemical, and Biological Evaluation of Fluorescent Chlorido[ N, N'-bis(methoxy/hydroxy)salicylidene-1,2-bis(4-methoxyphenyl)ethylenediamine]iron(III) Complexes as Anticancer Agents. J Med Chem 2023; 66:15916-15925. [PMID: 38013413 PMCID: PMC10726350 DOI: 10.1021/acs.jmedchem.3c01359] [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: 07/27/2023] [Revised: 11/02/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
The impact of methoxy and hydroxyl groups at the salicylidene moiety of chlorido[N,N'-bis(methoxy/hydroxy)salicylidene-1,2-bis(4-methoxyphenyl)ethylenediamine]iron(III) complexes was evaluated on human MDA-MB 231 breast cancer and HL-60 leukemia cells. Methoxylated complexes (C1-C3) inhibited proliferation, migration, and metabolic activity in a concentration-dependent manner following the rank order: C2 > C3 > C1. In particular, C2 was highly cytotoxic with an IC50 of 4.2 μM which was 6.6-fold lower than that of cisplatin (IC50 of 27.9 μM). In contrast, hydroxylated complexes C4-C6 were almost inactive up to the highest concentration tested due to lack of cellular uptake. C2 caused a dual mode of cell death, ferroptosis, and necroptosis, whereby at higher concentrations, ferroptosis was the preferred form. Ferroptotic morphology and the presence of ferrous iron and lipid reactive oxygen species proved the involvement of ferroptosis. C2 was identified as a promising lead compound for the design of drug candidates inducing ferroptosis.
Collapse
Affiliation(s)
- Astrid
Dagmar Bernkop-Schnürch
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI−Center
for Molecular Biosciences Innsbruck, CCB—Center for Chemistry
and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Donja Chavooshi
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI−Center
for Molecular Biosciences Innsbruck, CCB—Center for Chemistry
and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
- Immunobiology
and Stem Cell Laboratory, Department of Internal Medicine V (Hematology
and Oncology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Hubert Aaron Descher
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI−Center
for Molecular Biosciences Innsbruck, CCB—Center for Chemistry
and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Daniel Leitner
- Department
of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Heribert Talasz
- Biocenter,
Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Martin Hermann
- Department
of Anesthesiology and Critical Care Medicine, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
| | - Klaus Wurst
- Department
of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Stephan Hohloch
- Department
of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Ronald Gust
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI−Center
for Molecular Biosciences Innsbruck, CCB—Center for Chemistry
and Biomedicine, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Brigitte Kircher
- Immunobiology
and Stem Cell Laboratory, Department of Internal Medicine V (Hematology
and Oncology), Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Tyrolean
Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
| |
Collapse
|
4
|
Piñera-Avellaneda D, Buxadera-Palomero J, Ginebra MP, Rupérez E, Manero JM. Gallium-doped thermochemically treated titanium reduces osteoclastogenesis and improves osteodifferentiation. Front Bioeng Biotechnol 2023; 11:1303313. [PMID: 38144539 PMCID: PMC10748490 DOI: 10.3389/fbioe.2023.1303313] [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: 09/27/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Excessive bone resorption is one of the main causes of bone homeostasis alterations, resulting in an imbalance in the natural remodeling cycle. This imbalance can cause diseases such as osteoporosis, or it can be exacerbated in bone cancer processes. In such cases, there is an increased risk of fractures requiring a prosthesis. In the present study, a titanium implant subjected to gallium (Ga)-doped thermochemical treatment was evaluated as a strategy to reduce bone resorption and improve osteodifferentiation. The suitability of the material to reduce bone resorption was proven by inducing macrophages (RAW 264.7) to differentiate to osteoclasts on Ga-containing surfaces. In addition, the behavior of human mesenchymal stem cells (hMSCs) was studied in terms of cell adhesion, morphology, proliferation, and differentiation. The results proved that the Ga-containing calcium titanate layer is capable of inhibiting osteoclastogenesis, hypothetically by inducing ferroptosis. Furthermore, Ga-containing surfaces promote the differentiation of hMSCs into osteoblasts. Therefore, Ga-containing calcium titanate may be a promising strategy for patients with fractures resulting from an excessive bone resorption disease.
Collapse
Affiliation(s)
- David Piñera-Avellaneda
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Judit Buxadera-Palomero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Elisa Rupérez
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - José María Manero
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Barcelona East School of Engineering (EEBE), Technical University of Catalonia (UPC), Barcelona, Spain
- Barcelona Research Center in Multiscale Science and Engineering, EEBE, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| |
Collapse
|
5
|
Wang Y, Chen S, Wang C, Guo F. Nanocarrier-based targeting of metabolic pathways for endometrial cancer: Status and future perspectives. Biomed Pharmacother 2023; 166:115348. [PMID: 37639743 DOI: 10.1016/j.biopha.2023.115348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/10/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023] Open
Abstract
Cancer is the second-most lethal global disease, as per health reports, and is responsible for around 70% of deaths in low- and middle-income countries. Endometrial cancer is one of the emerging malignancies and has been predicted as a public health challenge for the future. Insulin resistance, obesity, and diabetes mellitus are the key metabolic factors that promote risks for the development of endometrial cancer. Various signaling pathways and associated genes are involved in the genesis of endometrial cancer, and any mutation or deletion in such related factors leads to the induction of endometrial cancer. The conventional way of drug delivery has been used for ages but is associated with poor management of cancer due to non-targeting of the endometrial cancer cells, low efficacy of the therapy, and toxicity issues as well. In this context, nanocarrier-based therapy for the management of endometrial cancer is an effective alternate choice that overcomes the problems associated with conventional therapy. In this review article, we highlighted the nanocarrier-based targeting of endometrial cancer, with a special focus on targeting various metabolic signaling pathways. Furthermore, the future perspectives of nanocarrier-based targeting of metabolic pathways in endometrial cancer were also underpinned. It is concluded that targeting metabolic signaling pathways in endometrial cancer via nanocarrier scaffolds is the future of pharmaceutical design for the significant management and treatment of endometrial cancer.
Collapse
Affiliation(s)
- Yichao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Siyao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Chunling Wang
- Medical Affairs Department, The Second Hospital of Jilin University, Changchun 130000, China
| | - Fengjun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
6
|
Ta N, Jiang X, Zhang Y, Wang H. Ferroptosis as a promising therapeutic strategy for melanoma. Front Pharmacol 2023; 14:1252567. [PMID: 37795022 PMCID: PMC10546212 DOI: 10.3389/fphar.2023.1252567] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
Malignant melanoma (MM) is the most common and deadliest type of skin cancer and is associated with high mortality rates across all races and ethnicities. Although present treatment options combined with surgery provide short-term clinical benefit in patients and early diagnosis of non-metastatic MM significantly increases the probability of survival, no efficacious treatments are available for MM. The etiology and pathogenesis of MM are complex. Acquired drug resistance is associated with a pool prognosis in patients with advanced-stage MM. Thus, these patients require new therapeutic strategies to improve their treatment response and prognosis. Multiple studies have revealed that ferroptosis, a non-apoptotic form of regulated cell death (RCD) characterized by iron dependant lipid peroxidation, can prevent the development of MM. Recent studies have indicated that targeting ferroptosis is a promising treatment strategy for MM. This review article summarizes the core mechanisms underlying the development of ferroptosis in MM cells and its potential role as a therapeutic target in MM. We emphasize the emerging types of small molecules inducing ferroptosis pathways by boosting the antitumor activity of BRAFi and immunotherapy and uncover their beneficial effects to treat MM. We also summarize the application of nanosensitizer-mediated unique dynamic therapeutic strategies and ferroptosis-based nanodrug targeting strategies as therapeutic options for MM. This review suggests that pharmacological induction of ferroptosis may be a potential therapeutic target for MM.
Collapse
Affiliation(s)
- Na Ta
- Department of Neurosurgery, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Xiaodong Jiang
- Department of Anatomy, College of Basic Medicine, Chifeng University Health Science Center, Chifeng, China
| | - Yongchun Zhang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Chifeng University, Chifeng, China
| | - Hongquan Wang
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
7
|
Xiang X, Gao J, Su D, Shi D. The advancements in targets for ferroptosis in liver diseases. Front Med (Lausanne) 2023; 10:1084479. [PMID: 36999078 PMCID: PMC10043409 DOI: 10.3389/fmed.2023.1084479] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Ferroptosis is a type of regulated cell death caused by iron overload and lipid peroxidation, and its core is an imbalance of redox reactions. Recent studies showed that ferroptosis played a dual role in liver diseases, that was, as a therapeutic target and a pathogenic factor. Therefore, herein, we summarized the role of ferroptosis in liver diseases, reviewed the part of available targets, such as drugs, small molecules, and nanomaterials, that acted on ferroptosis in liver diseases, and discussed the current challenges and prospects.
Collapse
Affiliation(s)
- Xiaohong Xiang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xiaohong Xiang
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danyang Su
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Doudou Shi
- Department of Geriatrics, The Ninth Hospital of Xi'an, Xi'an, Shaanxi, China
| |
Collapse
|
8
|
Zhang K, Gu X, Xia Y, Zhao X, Khoso Pervez A, Li S. MiR-129-3p regulates ferroptosis in the liver of Selenium-deficient broilers by targeting SLC7A11. Poult Sci 2022; 102:102271. [PMID: 36436380 PMCID: PMC9700304 DOI: 10.1016/j.psj.2022.102271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/12/2022] [Accepted: 10/15/2022] [Indexed: 11/23/2022] Open
Abstract
Selenium (Se) has been proven to be an essential trace element for organism. Se deficiency in poultry can cause widespread damage, such as exudative diathesis. The liver is not only the main organ of metabolism, but also one of the organs with high Se content in organism. Recent studies have shown that solute carrier family 7 member 11 (SLC7A11) plays a key role in the negative regulation of ferroptosis. In order to explore the mechanism of Se deficiency induces liver ferroptosis in broilers, and the role of microRNAs (miRNAs) in this process, we divided broilers into 2 groups: control group (0.2 mg/kg Se) and Se deficiency group (0.03 mg/kg Se). Hematoxylin-Eosin staining detected liver tissue damage in broilers. Predicted and verified the targeting relationship between miR-129-3p and SLC7A11 through miRDB and dual luciferase report experiments. The genes related to ferroptosis were detected by qRT-PCR and Western Blot. The results showed that the expression level of miR-129-3p mRNA in Se-deficient liver was significantly increased. To understand whether the miR-129-3p/SLC7A11 axis could involve in the process of ferroptosis, our further research showed that overexpression of miR-129-3p could reduce the expression of SLC7A11 and its downstream GCL, GSS, and GPX4, thereby inducing ferroptosis. These data indicates that miR-129-3p affected ferroptosis under Se deficiency conditions through the SLC7A11 pathway. Our research provides a new perspective for the mechanism of Se deficiency on the liver damage.
Collapse
Affiliation(s)
- Kaixin Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xuedie Gu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yu Xia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xiaochun Zhao
- Animal Disease Control and Prevention of Heilongjiang Province, Harbin 150069, China
| | - Ahmed Khoso Pervez
- Shaheed Benazir Bhutto, University of Veterinary and Animal Sciences, Sakrand, Pakistan
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| |
Collapse
|
9
|
Li C, Wu X, Zheng C, Xu S, Liu Y, Qin J, Fan X, Ye Y, Fei W. Nanotechnology-integrated ferroptosis inducers: a sharp sword against tumor drug resistance. J Mater Chem B 2022; 10:7671-7693. [PMID: 36043505 DOI: 10.1039/d2tb01350a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Presently, the biggest hurdle to cancer therapy is the inevitable emergence of drug resistance. Since conventional therapeutic schedules fall short of the expectations in curbing drug resistance, the development of novel drug resistance management strategies is critical. Extensive research over the last decade has revealed that the process of ferroptosis is correlated with cancer resistance; moreover, it has been demonstrated that ferroptosis inducers reverse drug resistance. To elucidate the development and promote the clinical transformation of ferroptosis strategies in cancer therapy, we first analyzed the roles of key ferroptosis-regulating molecules in the progression of drug resistance in-depth and then reviewed the design of ferroptosis-inducing strategies based on nanotechnology for overcoming drug resistance, including glutathione depletion, reactive oxygen species generation, iron donation, lipid peroxidation aggregation, and multiple-drug resistance-associated tumor cell destruction. Finally, the prospects and challenges of regulating ferroptosis as a therapeutic strategy for reversing cancer therapy resistance were evaluated. This review aimed to provide a comprehensive understanding for researchers to develop ferroptosis-inducing nanoplatforms that can overcome drug resistance.
Collapse
Affiliation(s)
- Chaoqun Li
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Xiaodong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Shanshan Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxi Liu
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Jiale Qin
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaoyu Fan
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
10
|
Iron Single-Atom nanocatalysts in response to tumor microenvironment for highly efficient Chemo-chemodynamic therapy. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
11
|
Gallium(III) Complex with Cloxyquin Ligands Induces Ferroptosis in Cancer Cells and Is a Potent Agent against Both Differentiated and Tumorigenic Cancer Stem Rhabdomyosarcoma Cells. Bioinorg Chem Appl 2022; 2022:3095749. [PMID: 35502218 PMCID: PMC9056256 DOI: 10.1155/2022/3095749] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/22/2022] [Indexed: 12/30/2022] Open
Abstract
In this work, gallium(III) complex with cloxyquin (5-chloro-8-quinolinol, HClQ) ligands is shown to effectively inhibit proliferation of rhabdomyosarcoma cells, the frequent, aggressive, and poorly treatable cancer of children. It offers striking selectivity to cancer cells compared to noncancerous human fibroblasts. The data reveal that the complex induces ferroptosis in rhabdomyosarcoma cells, likely due to interfering with iron metabolism. Importantly, it can kill both bulk and stem rhabdomyosarcoma cells. To the best of our knowledge, this is the first compound based on metal other than Fe capable of inducing ferroptosis in cancer cells.
Collapse
|
12
|
Shi Z, Zheng J, Tang W, Bai Y, Zhang L, Xuan Z, Sun H, Shao C. Multifunctional Nanomaterials for Ferroptotic Cancer Therapy. Front Chem 2022; 10:868630. [PMID: 35402376 PMCID: PMC8987283 DOI: 10.3389/fchem.2022.868630] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
Patient outcomes from the current clinical cancer therapy remain still far from satisfactory. However, in recent years, several biomedical discoveries and nanotechnological innovations have been made, so there is an impetus to combine these with conventional treatments to improve patient experience and disease prognosis. Ferroptosis, a term first coined in 2012, is an iron-dependent regulated cell death (RCD) based on the production of reactive oxygen species (ROS) and the consequent oxidization of polyunsaturated fatty acids (PUFAs). Many nanomaterials that can induce ferroptosis have been explored for applications in cancer therapy. In this review, we summarize the recent developments in ferroptosis-based nanomaterials for cancer therapy and discuss the future of ferroptosis, nanomedicine, and cancer therapy.
Collapse
Affiliation(s)
- Zhiyuan Shi
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jianzhong Zheng
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenbin Tang
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yang Bai
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Zhang
- School of Public Health, Xiamen Univerisity, Xiamen, China
| | - Zuodong Xuan
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Huimin Sun
- Central Laboratory, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Huimin Sun, ; Chen Shao,
| | - Chen Shao
- Department of Urology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Huimin Sun, ; Chen Shao,
| |
Collapse
|
13
|
Zhao Y, Zhang Z, Pan Z, Liu Y. Advanced bioactive nanomaterials for biomedical applications. EXPLORATION (BEIJING, CHINA) 2021; 1:20210089. [PMID: 37323697 PMCID: PMC10191050 DOI: 10.1002/exp.20210089] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Bioactive materials are a kind of materials with unique bioactivities, which can change the cellular behaviors and elicit biological responses from living tissues. Bioactive materials came into the spotlight in the late 1960s when the researchers found that the materials such as bioglass could react with surrounding bone tissue for bone regeneration. In the following decades, advances in nanotechnology brought the new development opportunities to bioactive nanomaterials. Bioactive nanomaterials are not a simple miniaturization of macroscopic materials. They exhibit unique bioactivities due to their nanoscale size effect, high specific surface area, and precise nanostructure, which can significantly influence the interactions with biological systems. Nowadays, bioactive nanomaterials have represented an important and exciting area of research. Current and future applications ensure that bioactive nanomaterials have a high academic and clinical importance. This review summaries the recent advances in the field of bioactive nanomaterials, and evaluate the influence factors of bioactivities. Then, a range of bioactive nanomaterials and their potential biomedical applications are discussed. Furthermore, the limitations, challenges, and future opportunities of bioactive nanomaterials are also discussed.
Collapse
Affiliation(s)
- Yu Zhao
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for New Organic MatterCollege of ChemistryNankai UniversityTianjinP. R. China
| | - Zhanzhan Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for New Organic MatterCollege of ChemistryNankai UniversityTianjinP. R. China
| | - Zheng Pan
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for New Organic MatterCollege of ChemistryNankai UniversityTianjinP. R. China
| | - Yang Liu
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical BiologyFrontiers Science Center for New Organic MatterCollege of ChemistryNankai UniversityTianjinP. R. China
| |
Collapse
|
14
|
Tao J, Li C, Zheng Y, Wang F, Zhang M, Wu X, Chen Y, Zeng Q, Chen F, Fei W. Biological protein mediated ferroptotic tumor nanotherapeutics. J Mater Chem B 2021; 9:9262-9284. [PMID: 34730601 DOI: 10.1039/d1tb01289d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ferroptosis, a cell death pathway involving iron-related generation of lipid hydroperoxides for achieving incredible tumor suppression, has reignited the hope of chemotherapy in tumor treatment in the past decade. With extensive research studies, various bioactive proteins and cellular pathways have been demonstrated to regulate the occurrence and development of ferroptosis. The gradually established ferroptotic regulatory network is conducive to find effective proteins from a holistic perspective and guides better designs for future ferroptotic tumor therapies. The first section of this review summarizes the recent advances in ferroptotic regulatory mechanisms of proteins and attempts to clarify their latent function in the ferroptotic regulatory network. Second, the existing protein-mediated ferroptotic tumor nanotherapeutic strategies were reviewed, including the protein-mediated iron supplement, cell membrane transporter inhibition, glutathione peroxidase 4 interference, glutathione depletion, bioenzyme-mediated reactive oxygen species generation, heat shock protein inhibition, and tumor-overexpressed protein-triggered drug release for ferroptotic therapy. Finally, the future expectations and challenges of ferroptotic tumor nanotherapeutics for clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Jiaoyang Tao
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chaoqun Li
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Yongquan Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Fengmei Wang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Xiaodong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Qingquan Zeng
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Fengying Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
15
|
Wu S, Li T, Liu W, Huang Y. Ferroptosis and Cancer: Complex Relationship and Potential Application of Exosomes. Front Cell Dev Biol 2021; 9:733751. [PMID: 34568341 PMCID: PMC8455874 DOI: 10.3389/fcell.2021.733751] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Cell death induction has become popular as a novel cancer treatment. Ferroptosis, a newly discovered form of cell death, features regulated, iron-dependent accumulation of lipid hydroperoxides. Since this word “ferroptosis” was coined, numerous studies have examined the complex relationship between ferroptosis and cancer. Here, starting from the intrinsic hallmarks of cancer and cell death, we discuss the theoretical basis of cell death induction as a cancer treatment. We review various aspects of the relationship between ferroptosis and cancer, including the genetic basis, epigenetic modification, cancer stem cells, and the tumor microenvironment, to provide information and support for further research on ferroptosis. We also note that exosomes can be applied in ferroptosis-based therapy. These extracellular vesicles can deliver different molecules to modulate cancer cells and cell death pathways. Using exosomes to control ferroptosis occurring in targeted cells is promising for cancer therapy.
Collapse
Affiliation(s)
- Shuang Wu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tianye Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Yongye Huang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
16
|
Liu Y, Chen Y, Fei W, Zheng C, Zheng Y, Tang M, Qian Y, Zhang X, Zhao M, Zhang M, Wang F. Silica-Based Nanoframeworks Involved Hepatocellular Carcinoma Theranostic. Front Bioeng Biotechnol 2021; 9:733792. [PMID: 34557478 PMCID: PMC8452863 DOI: 10.3389/fbioe.2021.733792] [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: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Silica-based nanoframeworks have been extensively studied for diagnosing and treating hepatocellular carcinoma (HCC). Several reviews have summarized the advantages and disadvantages of these nanoframeworks and their use as drug-delivery carriers. Encouragingly, these nanoframeworks, especially those with metal elements or small molecular drugs doping into the skeleton structure or modifying onto the surface of nanoparticles, could be multifunctional components participating in HCC diagnosis and treatment rather than functioning only as drug-delivery carriers. Therefore, in this work, we described the research progress of silica-based nanoframeworks involved in HCC diagnosis (plasma biomarker detection, magnetic resonance imaging, positron emission tomography, photoacoustic imaging, fluorescent imaging, ultrasonography, etc.) and treatment (chemotherapy, ferroptotic therapy, radiotherapy, phototherapy, sonodynamic therapy, immunotherapy, etc.) to clarify their roles in HCC theranostics. Further, the future expectations and challenges associated with silica-based nanoframeworks were highlighted. We believe that this review will provide a comprehensive understanding for researchers to design novel, functional silica-based nanoframeworks that can effectively overcome HCC.
Collapse
Affiliation(s)
- Yunxi Liu
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Chen
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weidong Fei
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yongquan Zheng
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Miao Tang
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Ying Qian
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Zhang
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengdan Zhao
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Zhang
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengmei Wang
- Department of Pharmacy, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|