Next Article in Journal
Spectroscopic Constants and Anharmonic Vibrational Frequencies of C(O)OC, c-C2O2 and Their Silicon-Containing Analogues
Previous Article in Journal
Structural and Spectroscopic Study of New Copper(II) and Zinc(II) Complexes of Coumarin Oxyacetate Ligands and Determination of Their Antimicrobial Activity
Previous Article in Special Issue
Facile Synthesis of P-Doped ZnIn2S4 with Enhanced Visible-Light-Driven Photocatalytic Hydrogen Production
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Progress in the Mechanism of the Effect of Fe3O4 Nanomaterials on Ferroptosis in Tumor Cells

1
Health Science Center, Ningbo University, Ningbo 315211, China
2
The First Affiliated Hospital of Ningbo University, Ningbo 315211, China
3
Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Nanning 530021, China
4
Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning 530021, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(11), 4562; https://doi.org/10.3390/molecules28114562
Submission received: 29 April 2023 / Revised: 24 May 2023 / Accepted: 30 May 2023 / Published: 5 June 2023

Abstract

:
Ferroptosis is a new form of iron-dependent programmed cell death discovered in recent years, which is caused by the accumulation of lipid peroxidation (LPO) and reactive oxygen species (ROS). Recent studies have shown that cellular ferroptosis is closely related to tumor progression, and the induction of ferroptosis is a new means to inhibit tumor growth. Biocompatible Fe3O4 nanoparticles (Fe3O4-NPs), rich in Fe2+ and Fe3+, act as a supplier of iron ions, which not only promote ROS production but also participate in iron metabolism, thus affecting cellular ferroptosis. In addition, Fe3O4-NPs combine with other techniques such as photodynamic therapy (PDT); heat stress and sonodynamic therapy (SDT) can further induce cellular ferroptosis effects, which then enhance the antitumor effects. In this paper, we present the research progress and the mechanism of Fe3O4-NPs to induce ferroptosis in tumor cells from the perspective of related genes and chemotherapeutic drugs, as well as PDT, heat stress, and SDT techniques.

1. Introduction

Global Statistics of 2020 [1] showed that there were an estimated 19.3 million new cancer cases and nearly 10 million cancer deaths, of which female breast cancer surpassed lung cancer as the most commonly diagnosed cancer with an estimated 2.3 million new cases (11.7%), followed closely by lung cancer (11.4%), colorectal cancer (10.0%), prostate cancer (7.3%), and stomach cancer (5.6%). Lung cancer remained the leading cause of cancer deaths with an estimated 1.8 million deaths (18%), followed by colorectal cancer (9.4%), liver cancer (8.3%), stomach cancer (7.7%), and female breast cancer (6.9%). The proliferation and apoptosis of corresponding tumor cells are closely related to tumor formation and progression. Thus, in the past few decades in oncology, scientific researchers have been devoted to the development of various apoptosis-related drugs to reverse the fate of non-senescent and over-proliferating tumor cells [2,3,4].
However, the therapeutic outcome is far from satisfactory due to the inherent or acquired apoptosis resistance of tumor cells [5]. Lung cancer, with the highest age-standardized incidence rate (ASIR) and age-standardized mortality rate (ASMR) worldwide, and with 2.1 million new cases and 1.8 million deaths every year, has a very high mortality rate [6,7,8,9]. Small cell lung cancer (SCLC) is one of the most malignant types of lung cancer. It has a short multiplication time, patients are prone to rapid drug resistance during treatment, and the disease deteriorates rapidly after recurrence. Currently, there is no effective second-line single-agent chemotherapy regimen other than topotecan [10]. In addition, the incidence and mortality of hepatocellular carcinoma (HCC) has rapidly increased worldwide [11]. HCC is an aggressive and highly malignant liver tumor with poor survival rates, and most patients are clinically diagnosed at an advanced stage [12]. The pathogenesis of the tumor has been of great interest to researchers; however, due to its diversity and complexity, it still has a long way to go despite the large amount of human and material resources invested every year [13,14,15,16,17]. In recent years, a new form of cell death different from the apoptotic mechanism, namely ferroptosis, has attracted attention.
Ferroptosis, first proposed in 2012 by Scott J. Dixon et al., is a unique form of iron-dependent non-apoptotic cell death triggered by the oncogenic RAS-selective lethal small molecule Erastin [18]. Early studies found that cysteine is required for the growth and death inhibition of cells, and the absence of cystine in the culture medium resulted in the death of human fibroblasts due to glutathione (GSH) depletion. Extracellular and intracellular cysteine are required to maintain GSH biosynthesis and inhibit mammalian cell death, which could be prevented and treated with the lipophilic antioxidant (α-tocopherol) and iron chelators (deferoxamine) [19]. In the following years, with the further study of the mechanism of ferroptosis, it has gradually become a promising cancer treatment.
Compared with other metal nanomaterials [20,21,22,23], Fe3O4-NPs are one of the few FDA-approved nanomaterials for clinical use, which has simple preparation [24,25,26], safe and stable chemical properties, and good biocompatibility [27,28,29]. Moreover, it has been widely used in pathogen detection [30,31,32,33,34], tumor diagnosis and treatment [35,36,37], gene mutation analysis [38,39,40,41,42], targeted drug delivery [43,44,45], and nuclear magnetic resonance imaging (MRI) [46,47,48]. In addition, recent studies have shown that Fe3O4-NPs, due to their rich Fe2+ and Fe3+ contents, can affect the iron metabolism of tumor cells, increase local ROS levels in tumor tissues, participate in or even induce the occurrence of ferroptosis in tumor cells, and then be able to inhibit or kill tumor cells. In this paper, the research progress and mechanism of Fe3O4-NP-induced ferroptosis in tumor cells were reviewed.

2. Ferroptosis

Ferroptosis, a new form of cell death discovered in recent years, is an iron-dependent non-apoptotic form of cell death characterized by the accumulation of intracellular ROS [18]. This modality disrupts the redox homeostasis of cells and has a great potential to kill cancer cells [49,50]. Studies on the mechanisms of ferroptosis have focused on lipid peroxidation (LPO) and iron loading. Under normal circumstances, ROS is mainly produced by normal metabolism in mitochondria and is essential for cell signaling and tissue homeostasis [51]. Polyunsaturated fatty acid (PUFA) peroxidation induced by ROS is the main cause of ferroptosis. Furthermore, ferroptosis is regulated by a complex interaction between cellular redox homeostasis, lipid metabolism, and iron metabolism [52]. In terms of cell morphology, cells undergoing ferroptosis show some specific changes, such as the appearance of smaller mitochondria than normal, wrinkled mitochondrial membranes, reduced or absent mitochondrial cristae, and fragmented extracellular membrane [53]. The biochemical features of ferroptosis are cystine deficiency, GSH depletion, and glutathione peroxidase 4 (GPX4) inactivation [54]. However, the underlying mechanisms have not been fully elucidated.
The main cause of ferroptosis is the change of cell metabolism, which leads to the accumulation of LPO and ROS in the cells. According to existing research reports, the changes in cell metabolic pathways caused by ferroptosis mainly include the following aspects (Figure 1):
(1)
GSH synthesis pathway is inhibited and LPO accumulates, thus inducing ferroptosis in cells [55].
(2)
Iron metabolism is altered. Iron is a redox-active metal involved in ROS formation and LPO diffusion, and a rising iron level could increase cellular susceptibility to iron-dependent cell death [56]. The accepted explanation today is that Fe2+ can transfer electrons to intracellular oxygen, and then react with intracellular lipids to form LPO, which further induces ferroptosis [57,58]. Iron metabolism genes and iron metabolism regulation genes play a key role in intracellular system iron homeostasis. For example, the gene of Iron Responsive Element Binding Protein 2 (IREB2) is a key player in the Erastin-induced ferroptosis of HT-1080 fibrosarcoma cells and Calu-1 lung cancer cells [59]. Thus, intracellular iron overload is critical for ferroptosis [60].
(3)
ROS metabolic pathway effects. This pathway also plays an important role in ferroptosis. Cytosolic cystine/glutamate transport receptor (System Xc-) and voltage-dependent anion channels (VDACs) [18] in the outer mitochondrial membrane, GPX4 and ferroptosis suppressor protein 1 (FSP1) ferroptosis-related proteins [61,62], and p62/keap1/Nrf2 [63], p53-related pathway [64,65], and ACSL4/LPCTA3/LOX [66] ferroptosis-related pathways play their roles in regulating ferroptosis by affecting ROS metabolism pathways [67].

3. Mechanism of the Effect of Fe3O4-NPs on Ferroptosis in Tumor Cells

Research on tumor detection, diagnosis, and treatment methods has been a popular topic, and the development of nanotechnology has provided new strategies in this field [68,69,70,71,72,73,74,75]. Fe2+ and Fe3+, rich in Fe3O4-NPs, contain abundant lone-pair electrons and can form coordination bonding with organic molecules. Thus, Fe3O4-NPs can easily realize surface modification and surface drug loading. After surface modification, Fe3O4-NPs are more difficultly encapsulated by plasma proteins and can avoid the phagocytosis of the reticuloendothelial system, reduce their clearance rate, and prolong the circulation time in blood [76,77,78,79].
Moreover, both Fe2+ and Fe3+ can react with hydrogen peroxide to generate ROS, catalyze the occurrence of the Fenton reaction, and then induce ferroptosis in cells [57]. Furthermore, it can enhance immunotherapy, which has attracted much more attention. Overall, iron-based nanoparticles as an emerging ferroptosis inducer can enhance iron ion and ROS levels. For example, Fe3O4-based nanoparticles prepared by Song et al. can generate ROS, induce intracellular oxidative stress, and accelerate the Fenton reaction to increase the local concentration of Fe2+, Fe3+, and H2O2 to kill cancer cells (Figure 2) [80].
Tian et al. [81] synthesized Fe3O4-NPs with different sizes (with an average size of 2, 4, 10, and 100 nm) and evaluated their antitumor effects (Figure 3). The experimental results showed that, compared to larger Fe3O4-NPs, the ultra-small (<5 nm) Fe3O4-NPs could accumulate in the nucleus and had a higher efficiency of •OH and ROS generation and a higher ability of cytokines secretion. Additionally, the ultra-small Fe3O4-NPs were more efficient in enhancing the toxicity of H2O2 due to the rapid release of Fe2+. Nevertheless, 10 nm Fe3O4-NPs displayed the best antitumor effect in vivo. It shows that selecting the right size of Fe3O4-NPs is more effective in treating tumors.
Ferulic acid glycerol, a nano-Fe supplementation, was found to have antileukemic effects under low levels of iron transport protein (FPN) in leukemic cells by inducing ferroptosis in vitro and in vivo [82]. Leukemic cells are unable to export intracellular Fe2+ produced by methyl ferulate, which can cause an increase in ROS levels through the Fenton reaction and then affect leukemic cells [83]. In addition, Zanganeh et al. showed that triglyceride ferulate has an intrinsic therapeutic effect on the growth of liver and lung metastases in early breast cancer and lung cancer [84].

3.1. Effect of Fe3O4-NPs on the Expression of Ferroptosis-Related Genes

In addition to lipid peroxidation [85,86,87], the role of genes involved in ferroptosis should also not be ignored. It has been shown that p53 activation is required for ferroptosis in cancer cells [88]. p53 can mediate ferroptosis in fibroblasts, osteosarcoma cells, and breast cancer cells through the transrepression of SLC7A11 [64,88]. It is the first piece of evidence of ferroptosis-induced p53 gene expression. p53 plays a dual role in ferroptosis. On the one hand, p53, as a tumor suppressor, down-regulates SLC7A11, thereby inhibiting the cellular uptake of cysteine, increasing cellular LPO and ultimately leading to ferroptosis. On the other hand, p53 inhibited Erastin-induced ferroptosis by blocking dipeptidyl peptidase 4 (DPP4) activity in human colorectal cancer (CRC) [89]. Fe3O4-NPs could promote the expression of p53 and acetylated p53 protein and accelerate their translocation from cytoplasm to nucleus in human umbilical vein endothelial cells; silencing p53 could up-regulate SLC7A11 and GPX4 mRNA expression levels (Figure 4) [90]. For macrophages, RNA sequencing results showed that Fe3O4-NPs induced ferroptosis through up-regulating p53 gene expression and down-regulating SLC7A11 gene expression [91]. In ovarian cancer cells incubated with superparamagnetic iron oxide, iron repletion could promote p53 expression [92,93] and up-regulated p53 promoted ferroptosis. These results suggest that Fe3O4-NPs could promote the expression of p53, which is involved in the expression of SLC7A11 and GPX4 to regulate ferroptosis.
In addition, there are other genes that play an equally important role in ferroptosis such as BRCA1-Associated Protein 1 (BAP1). BAP1, as a tumor suppressor, promotes ferroptosis and inhibits cancer cell growth by inhibiting SLC7A11 [94]. The mechanism may be that BAP1 suppresses SLC7A11 transcription by reducing histone 2A ubiquitination (H2Aub) on the SLC7A11 gene. Gao et al. [83] developed a cancer treatment named gene interference ferroptosis therapy (GIFT) by combining a DMP-controlled gene interference tool with DMSA-coated Fe3O4-NPs. GIFT consists of a gene interference vector (GIV) and Fe3O4-NPs, and DMP, an NF-κB-specific promoter, consisting of an NF-κB decoy and a minimal promoter. In a variety of cancer cells treated with Fe3O4-NPs, the expression of FPN and LCN2 (iron metabolism genes) was selectively down-regulated by Cas13a or microRNA controlled by the NF-κB-specific promoter and ferroptosis was significantly induced, but the same treatment had little effect on normal cells.
The Beclin1/ATG5-dependent autophagy pathway is also closely related to ferroptosis induced by Fe3O4-NPs. Wen et al. [95] demonstrated that down-regulated Beclin1/ATG5 could significantly inhibit the ferroptosis induced by ultra-small iron oxide nanoparticles (USIONPs), while overexpressed Beclin1/ATG5 could significantly enhance ferroptosis. It suggests that USIONP-induced ferroptosis is regulated through the Beclin1/ATG5-dependent autophagy pathway, which could lead to the degradation of USIONPs, the release of iron ions, and the accumulation of ROS and LPO, and ultimately induce cellular ferroptosis.

3.2. Fe3O4-NPs Enhance the Sensitivity of Tumor Cells to Anticancer Drugs

There is growing evidence that the induction of ferroptosis enhances susceptibility to anticancer drugs [96,97]. Inducing ferroptosis with Erastin and RSL-3 has been reported to enhance the anticancer effects of cisplatin and reduce the resistance of anticancer drugs by inhibiting System Xc- in lung cancer, CRC, ovarian cancer, and pancreatic ductal adenocarcinoma [98]. Fe3O4-NPs provide new ideas for improving the sensitivity of tumors to chemotherapeutic drugs by inducing ferroptosis, while providing new methods for in vitro high-sensitive detection [17,68,99,100,101]. Gao et al. [102] prepared a peptide carrier (Pt&Fe3O4@PP) (Figure 5) encapsulated with the anticancer drug cisplatin (Pt drug) and Fe3O4-NPs. Cisplatin, as an inducer of ferroptosis and apoptosis for NSCLC A549 cells, plays an important role in the mechanism of ferroptosis through reducing GSH depletion and inducing GPX4 inactivation [103]. The tumor microenvironment (TME) triggers the release of Pt and Fe2+/Fe3+, which in turn induces an intracellular cascade reaction to produce sufficient ·OH for ferroptosis treatment. In addition, the release of Pt could induce apoptosis in tumor cells, and Fe3O4-NPs can be used for T2-weighted imaging of tumors. It was further found that the combination of cisplatin and class I FIN Erastin had a significant synergistic effect on its antitumor activity. Liu et al. [104] established a novel drug delivery system (Fe3O4-siPD-L1@M-BV2) targeting glioblastoma multiforme (GBM). This system could induce ferroptosis in drug-resistant GBM cells and matured dendritic cells (DC), increase the proportion of M1 and M2 microglia in drug-resistant GBM tissues, and then inhibit the in situ growth and prolong the survival time of drug-resistant GBM mice. PD-1/PD-L1 inhibitors could activate T cells to secrete γ-interferons [105]. These γ-interferons inhibit cysteine transporters, thereby preventing cysteine uptake and reducing glutathione synthesis in cancer cells, and finally enhancing ferroptosis. Both in combination with Fe3O4-NPs and with the drug itself, the drug systems have a facilitative effect on the onset of ferroptosis by reducing glutathione in cancer cells.
Drug-loaded microspheres, as a commonly used clinical chemoembolic agent, suffer from inhomogeneous particle size and unstable efficacy. Chen et al. [106] introduced Fe3O4-NPs into the chemoembolic agent system to prepare gelatin microspheres co-loaded with doxorubicin (ADM/Fe3O4-MS) to improve its anti-liver cancer effect. Ferroptosis was also involved in the process of tumor cell death, in which GPX4, a marker of ferroptosis, was significantly reduced and ACSL4 was significantly increased. Meanwhile, ferroptosis inhibitors reversed the killing effect induced by ADM/Fe3O4-MS plus microwave irradiation. This study demonstrates that Fe3O4-NPs can significantly improve the overall efficacy of drug-loaded microspheres, which has promising applications especially in tumor chemoembolization [107].
Ferroptosis plays an important role in the treatment of triple-negative breast cancer (TNBC) cells. Yao et al. [108] prepared a novel ferroptosis nanodrug by loading simvastatin (SIM) into amphoteric polymer-coated magnetic nanoparticles (Fe3O4@PCBMA) to improve the therapeutic effect of TNBC. SIM could inhibit the expression of HMG-CoA reductase (HMGCR) and down-regulated the mevalonate (MVA) and GPX4 pathways, and consequently induced ferroptosis in cancer cells. Fe3O4-NPs serve as a good carrier to deliver antitumor drugs to tumor tissues through passive targeting. Based on this study, it is known that an Fe3O4-NP nanosystem can overcome the drug resistance caused by apoptotic drugs in tumor cells in clinical practice.
Wu et al. [109] developed a nanomedicine with both nanocatalytic and glutaminase inhibitor properties, which was made by integrating ultrasmall Fe3O4-NPs and CB-839 into dendritic mesoporous silica nanoparticles and then etched with Mn (DFMC) (Figure 6). The Fe2+ and Mn2+ in DFMC exerted Fenton/Fenton-like activity and could effectively catalyze the decomposition of H2O2 to be highly toxic ·OH under acidic conditions for tumor therapy. The GSH depletion ability of DFMC not only consumes existing GSH, but also prevents the synthesis of GSH, which weakens the GSH-related antioxidant defense system (ADS), thereby enhancing the sensitivity of ROS-mediated tumor catalytic therapy and oxaliplatin chemotherapy, and finally achieving higher tumor clearance rate.

3.3. Fe3O4-NPs Can Enhance the Efficacy of Drugs or Synergize with Them to Promote Ferroptosis

In one study, magnetic nanoparticles (MNP) were coated with the zwitterionic polymer Poly (2-methacryloyloxyethyl phosphorylcholine) (PMPC) and then loaded with sorafenib (SRF) to obtain the drug-loaded nanoparticles composite MNP@PMPC-SRF [110].
Fe3O4-NPs can also improve the anticancer efficiency of artemisinin (ART). The PFH/ART@PLGA/Fe3O4-EFA constructed by Wang et al. [110] was exposed to Low Intensity Focused Ultrasound (LIFU) irradiation to induce perfluorohexane (PFH) phase transition and nanoparticle collapse, which promoted the release of ART and Fe3O4 in vitro. The efficiency of this approach is attributed to the synergistic effect of intracellular Fe2+ and ART, which play a key role in the induction of ferroptosis in tumor cells by promoting ROS production in vitro.

4. Fe3O4-NPs in Combination with PDT, Heat Stress, and SDT Further Induced Ferroptosis in Tumor Cells

The combination of Fe3O4-NPs with other technologies further exploits their advantages and expands the scope of application [111,112,113,114,115,116]. The results of established studies have shown that Fe3O4-NPs are of great interest as a good cellular ferroptosis inducer, especially in the field of tumor therapy. However, current iron-based nanoparticles, due to their low ROS generation efficiency, require synergistic use with other therapeutic approaches or high dose application to achieve effective treatment [117].

4.1. Synergy of Photodynamic Therapy (PDT)

PDT is a novel tumor treatment method. However, the efficacy of PDT is greatly affected by oxygen concentration [118], which applied alone is not satisfactory. Fe3O4-NPs are highly biocompatible in vivo. When they enter cancer cells by endocytosis, they can be effectively released into Fe2+ and Fe3+ in the acidic environment of cancer cells, thus enhancing the Fenton reaction [119,120]. The Fenton reaction can produce ·OH and O2, increase the oxygen concentration in tumor cells, enhance the PDT effect, and then realize the treatment of cancers.
Chen et al. [121] designed a nanosystem coated with the FDA-approved poly (lactic-co-glycolic acid) (PLGA) containing Fe3O4-NPs and chlorin E6 (Ce6) for synergistic ferroptosis-photodynamic anticancer therapy (Figure 7). The nanosystem can dissociate in acidic TME to release Fe2+/Fe3+ and Ce6. Fe2+/Fe3+ may react with the excess intracellular H2O2 to trigger the Fenton reaction, which allows intracellular ROS accumulation and LPO generation, providing the necessary conditions for the onset of ferroptosis. Meanwhile, Ce6 could also increase the generation and accumulation of ROS under laser irradiation, and then provide PDT to further promote cellular ferroptosis. It demonstrates that the synergistic effect of PDT and ferroptosis induced by Fe3O4-NPs has an efficient effect on tumor suppression.
Ultrafine Fe3O4@PGL complex nanoparticles, designed by Liang et al. [122], were formed by amphiphilic porphyrin-grafted lipid (PGL) self-assembly on the hydrophobic surface of ultrasmall Fe3O4-NPs. This complex could not only trigger PDT under laser action, but also induce a large amount of ROS through the activity of tumor-associated macrophages (TAMs). Experimental results confirmed that most cells followed a PDT-based cell death pathway, and Fe3O4-mediated ferroptosis can further improve the therapeutic efficiency, showing a strong synergistic effect. Under the synergistic effect of ferroptosis and PDT, tumor cells regressed [123].
In summary, Fe3O4-NPs in an acidic environment (such as TME and lysosomes) can dissociate and then release Fe2+/Fe3+ diffusing into the cytoplasm, which accelerates the Fenton reaction, promotes ferroptosis, and leads to intracellular ROS accumulation and LPO generation [124]. PDT can also promote ROS production and accumulation, which in turn promotes the occurrence of ferroptosis. Therefore, the combination of Fe3O4-NPs and PDT can effectively promote ferroptosis.

4.2. Metabolism Modulation by Heat Stress

Peptide-modified and 1H-perfluoropentane (1H-PFP)-coated Fe3O4-NPs (GBP@Fe3O4) have been rationally designed by researchers to propose a hypothesis of heat-triggered tumor-specific ferroptosis (Figure 8) [125]. When a GBP@Fe3O4 complex is irradiated by an 808 nm laser, the phase transition of 1H-PFP could be triggered by localized heat (45 °C), which leads to an abrupt release of Fe3O4-NPs in situ, and then generates intense ROS through the Fenton reaction in the TME. Oxidative damage, antioxidant inhibitory response, and the unique reprogramming of lipid metabolism during heat stress are amplified to induce tumor ferroptosis and achieve sufficient antitumor effects. Biocompatible Fe3O4-NPs act as a supplier of iron ions and can not only promote ROS production, but also participate in iron metabolism associated with ferroptosis [126]. Moderate heat stress has no significant effect on cellular ROS production, but it can significantly amplify Fe3O4-NP-induced oxidative stress and induce apoptosis in cancer cells [125]. Therefore, moderate heat stress acts synergistically with Fe3O4-NPs to weaken the self-defense response of tumors and affect their metabolism, thus regulating their mode of death.
A plate-like Bi2Se3-Fe3O4/Au (BFA) nanoplatform was designed for clinical diagnosis and therapy [127], which can increase the rate of the Fenton reaction to enhance ferroptosis therapy with active–passive targeting. This platform benefits from the internal synergistic effects of Fe3O4-NPs and Au-NPs, as well as external near-infrared light (NIR)-mediated hyperthermia, and could promote hydroxyl radical (·OH) production to enhance intracellular oxidative stress and further induce ferroptosis by inactivating GPX4. At the same time, BFA-NPs can be used as an effective diagnostic agent for photoacoustic (PA), magnetic resonance (MR), and X-ray imaging to guide the synergistic treatment of photothermal-ferroptosis. CdSe/Fe3O4 nanoplatforms coated with multi-mode polymers had both optical and magnetic functions [128,129]. They co-wrapped with CdSe quantum dots and Fe3O4-NPs, and it was demonstrated for the first time that they may have biomedical applications based on two-photon and magnetothermal therapies such as bioimaging and anticancer therapy. However, co-wrapping limits CdSe/Fe3O4 nanoplatforms inducing iron-dependent and LPO-mediated ferroptosis. Chen et al. [106] confirmed the synergistic effect of hyperthermia and Fe3O4-NPs from another aspect. They designed an ADM/Fe3O4-MS complex to enhance their antitumor effects by activating ferroptosis under microwave heating.
Extensive studies have shown that thermotherapy is an effective sensitizer of tumor chemoresistancy, which can greatly improve the effectiveness of chemotherapy [130] and enhance the antitumor effect of chemotherapeutic agents [131]. Therefore, the introduction of hyperthermia into the chemoembolization system could significantly improve the efficacy of transcatheter arterial chemoembolization (TACE). In conclusion, moderate heat, rather than a heat source, is essential for the induction of ferroptosis in combination therapy [125].

4.3. Promotion of Sonodynamic Therapy (SDT)

Recently, researchers have proposed a combination therapy strategy for glioma based on a non-invasive blood-brain barrier (BBB) opening, biomimetic sound-therapy-system-mediated SDT, and ferroptosis [132]. In this study, multifunctional homologous tumor-targeting biomimetic nanoparticles (PIOC@CM-NPs) coated with Fe3O4-NPs and Ce6 were constructed as a nanoultrasound sensitizer, which combined the biomimetic nanoacoustic-sensitizer-mediated SDT with ferroptosis to achieve the effect of synergistic treatment of glioma. It demonstrated that the glioma C6 cell membrane on the surface of nanoparticles allowed selective accumulation of nanosensitized agents in tumors by homologous targeting in vitro. After effective internalization in C6 cells, PIOC@CM-NPs can significantly improve ROS levels and deplete GSH upon ultrasound irradiation, resulting in a loss of GPX4 activity, thereby promoting SDT and ferroptosis to kill glioma C6 cells.

5. Conclusions and Perspectives

Ferroptosis, a new iron-dependent programmed death discovered in recent years, has received increasing attention as a unique mechanism of occurrence and resistance, unlike apoptosis, cell necrosis, and cell autophagy. The activation of ferroptosis is widely recognized as a new target for drug discovery, and an increasing number of small molecule compounds have been identified and characterized to induce ferroptosis directly or indirectly by targeting iron metabolism and LPO. However, there are relatively few reports on the relationship between nanomaterials and cell ferroptosis, especially Fe3O4-NPs, which are rich in Fe2+ and Fe3+ and have unique advantages in inducing ferroptosis; however, the specific role and influencing mechanism still requires further research. Although there are many in vitro assays available to detect ferroptosis, such as by measuring cell viability, iron content, and ROS levels, confirming the presence of ferroptosis in vivo is more difficult. Ferroptosis is a double-edged sword, and we need to fully investigate the potential toxicities of inducers or inhibitors of key proteins and pathways of ferroptosis to ensure tumor-specific triggering of the Fenton response and avoid off-target toxicity to normal tissues causing carcinogenesis or other diseases [133,134].

Author Contributions

Conceptualization, X.M.; writing and software, Y.W.; software, X.B.; review and editing, X.W. and X.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Natural Science Foundation of Ningbo (2022J130), Open in Project of Guangxi Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University) (GKE-KF202009), and General Scientific Research Project of Zhejiang Education Department (Y202146345).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Hassannia, B.; Vandenabeele, P.; Vanden Berghe, T. Targeting Ferroptosis to Iron Out Cancer. Cancer Cell 2019, 35, 830–849. [Google Scholar] [CrossRef] [PubMed]
  3. Li, F.; Wang, Z.; Huang, Y.; Xu, H.; He, L.; Deng, Y.; Zeng, X.; He, N. Delivery of PUMA Apoptosis Gene Using Polyethyleneimine-SMCC-TAT/DNA Nanoparticles: Biophysical Characterization and In Vitro Transfection Into Malignant Melanoma Cells. J. Biomed. Nanotechnol. 2015, 11, 1776–1782. [Google Scholar] [CrossRef] [PubMed]
  4. Fu, J.; Dang, Z.; Deng, Y.; Lu, G. Regulation of c-Myc and Bcl-2 induced apoptosis of human bronchial epithelial cells by zinc oxide nanoparticles. J. Biomed. Nanotechnol. 2012, 8, 669–675. [Google Scholar] [CrossRef]
  5. Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef] [PubMed]
  6. Li, W.; Liu, Y.; Li, Z.; Shi, Y.; Deng, J.; Bai, J.; Ma, L.; Zeng, X.; Feng, S.; Ren, J.; et al. Unravelling the Role of LncRNA WT1-AS/miR-206/NAMPT Axis as Prognostic Biomarkers in Lung Adenocarcinoma. Biomolecules 2021, 11, 203. [Google Scholar] [CrossRef]
  7. Li, W.; Jia, M.; Deng, J.; Wang, J.; Lin, Q.; Tang, J.; Zeng, X.; Cai, F.; Ma, L.; Su, W.; et al. Down-regulation of microRNA-200b is a potential prognostic marker of lung cancer in southern-central Chinese population. Saudi J. Biol. Sci. 2019, 26, 173–177. [Google Scholar] [CrossRef]
  8. Li, W.; Jia, M.; Wang, J.; Lu, J.; Deng, J.; Tang, J.; Liu, C. Association of MMP9-1562C/T and MMP13-77A/G Polymorphisms with Non-Small Cell Lung Cancer in Southern Chinese Population. Biomolecules 2019, 9, 107. [Google Scholar] [CrossRef] [Green Version]
  9. Yang, S.; Guo, H.; Wei, B.; Zhu, S.; Cai, Y.; Jiang, P.; Tang, J. Association of miR-502-binding site single nucleotide polymorphism in the 3′-untranslated region of SET8 and TP53 codon 72 polymorphism with non-small cell lung cancer in Chinese population. Acta Biochim. Biophys. Sin. 2014, 46, 149–154. [Google Scholar] [CrossRef] [Green Version]
  10. Xing, H.; Zhang, J.; Ge, F.J.; Yu, X.H.; Bian, H.M.; Zhang, F.L.; Fang, J. Analysis of the Efficacy of Irinotecan in the Second-line Treatment of Refractory and Relapsed Small Cell Lung Cancer. Chin. J. Lung Cancer 2021, 24, 167–172. [Google Scholar] [CrossRef]
  11. Singal, A.G.; Lampertico, P.; Nahon, P. Epidemiology and surveillance for hepatocellular carcinoma: New trends. J. Hepatol. 2020, 72, 250–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Eggert, T.; Greten, T.F. Current Standard and Future Perspectives on Non-Surgical Therapy for Hepatocellular Carcinoma. Digestion 2017, 96, 1–4. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, J.; Zhang, S.; Wang, Y.; Xie, R.; Liu, L.; Deng, Y. In Vivo Self-Assembly Based Cancer Therapy Strategy. J. Biomed. Nanotechnol. 2020, 16, 997–1017. [Google Scholar] [CrossRef]
  14. Liu, M.; Yu, X.C.; Chen, Z.; Yang, T.; Yang, D.D.; Liu, Q.Q.; Du, K.K.; Li, B.; Wang, Z.F.; Li, S.; et al. Aptamer selection and applications for breast cancer diagnostics and therapy. J. Nanobiotechnol. 2017, 15, 81. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, M.; Xi, L.; Tan, T.; Jin, L.; Wang, Z.F.; He, N.Y. A novel aptamer-based histochemistry assay for specific diagnosis of clinical breast cancer tissues. Chin. Chem. Lett. 2021, 32, 1726–1730. [Google Scholar] [CrossRef]
  16. Xie, H.; Di, K.L.; Huang, R.R.; Khan, A.; Xia, Y.Y.; Xu, H.P.; Liu, C.; Tan, T.T.; Tian, X.Y.; Shen, H.; et al. Extracellular vesicles based electrochemical biosensors for detection of cancer cells: A review. Chin. Chem. Lett. 2020, 31, 1737–1745. [Google Scholar] [CrossRef]
  17. Li, Z.Y.; Wang, J.H.; Yang, H.W.; Chen, S.Q.; Ma, G.J.; Zhang, X.M.; Zhu, M.; Yu, J.; Singh, R.; Zhang, Y.Y.; et al. Ultrasensitive Detection of Gastric Cancer Plasma MicroRNAs via Magnetic Beads-Based Chemiluminescent Assay. J. Biomed. Nanotechnol. 2017, 13, 1272–1280. [Google Scholar] [CrossRef]
  18. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [Green Version]
  19. Bannai, S.; Tsukeda, H.; Okumura, H. Effect of antioxidants on cultured human diploid fibroblasts exposed to cystine-free medium. Biochem. Biophys. Res. Commun. 1977, 74, 1582–1588. [Google Scholar] [CrossRef]
  20. Magesa, F.; Wu, Y.Y.; Dong, S.; Tian, Y.L.; Li, G.L.; Vianney, J.M.; Buza, J.; Liu, J.; He, Q.G. Electrochemical Sensing Fabricated with Ta2O5 Nanoparticle-Electrochemically Reduced Graphene Oxide Nanocomposite for the Detection of Oxytetracycline. Biomolecules 2020, 10, 110. [Google Scholar] [CrossRef] [Green Version]
  21. Yang, G.J.; Huang, H.; Xiao, Z.Q.; Zhang, C.X.; Guo, W.F.; Ma, T.T.; Ma, L.; Chen, Z.; Deng, Y. A Novel Strategy for Liquid Exfoliation of Ultrathin Black Phosphorus Nanosheets. J. Biomed. Nanotechnol. 2020, 16, 548–552. [Google Scholar] [CrossRef] [PubMed]
  22. He, Q.G.; Tian, Y.L.; Wu, Y.Y.; Liu, J.; Li, G.L.; Deng, P.H.; Chen, D.C. Electrochemical Sensor for Rapid and Sensitive Detection of Tryptophan by a Cu2O Nanoparticles-Coated Reduced Graphene Oxide Nanocomposite. Biomolecules 2019, 9, 176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. He, Q.G.; Liu, J.; Liu, X.P.; Li, G.L.; Deng, P.H.; Liang, J. Manganese dioxide Nanorods/electrochemically reduced graphene oxide nanocomposites modified electrodes for cost-effective and ultrasensitive detection of Amaranth. Colloids Surf. B-Biointerfaces 2018, 172, 565–572. [Google Scholar] [CrossRef]
  24. Liu, J.; Dong, S.; He, Q.G.; Yang, S.C.; Xie, M.; Deng, P.H.; Xia, Y.H.; Li, G.L. Facile Preparation of Fe3O4/C Nanocomposite and Its Application for Cost-Effective and Sensitive Detection of Tryptophan. Biomolecules 2019, 9, 245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jiang, H.R.; Zeng, X.; Xi, Z.J.; Liu, M.; Li, C.Y.; Li, Z.Y.; Jin, L.; Wang, Z.F.; Deng, Y.; He, N.Y. Improvement on Controllable Fabrication of Streptavidin-Modified Three-Layer Core-Shell Fe3O4@SiO2@Au Magnetic Nanocomposites with Low Fluorescence Background. J. Biomed. Nanotechnol. 2013, 9, 674–684. [Google Scholar] [CrossRef]
  26. Ma, C.; Li, C.Y.; He, N.Y.; Wang, F.; Ma, N.N.; Zhang, L.M.; Lu, Z.X.; Ali, Z.; Xi, Z.J.; Li, X.L.; et al. Preparation and Characterization of Monodisperse Core-Shell Fe3O4@SiO2 Microspheres and Its Application for Magnetic Separation of Nucleic Acids from E. coli BL21. J. Biomed. Nanotechnol. 2012, 8, 1000–1005. [Google Scholar] [CrossRef]
  27. Fan, L.; Liu, H.; Gao, C.X.; Zhu, P.Z. Facile synthesis and characterization of magnetic hydroxyapatite/Fe3O4 microspheres. Mater. Lett. 2022, 313, 131648. [Google Scholar] [CrossRef]
  28. Liu, X.L.; Zhang, H.; Chang, L.; Yu, B.Z.; Liu, Q.Y.; Wu, J.P.; Miao, Y.Q.; Ma, P.; Fan, D.D.; Fan, H.M. Human-like collagen protein-coated magnetic nanoparticles with high magnetic hyperthermia performance and improved biocompatibility. Nanoscale Res. Lett. 2015, 10, 28. [Google Scholar] [CrossRef] [Green Version]
  29. Zhao, Y.; Fan, T.T.; Chen, J.D.; Su, J.C.; Zhi, X.; Pan, P.P.; Zou, L.; Zhang, Q.Q. Magnetic bioinspired micro/nanostructured composite scaffold for bone regeneration. Colloids Surf. B-Biointerfaces 2019, 174, 70–79. [Google Scholar] [CrossRef]
  30. He, L.; Yang, H.W.; Xiao, P.F.; Singh, R.; He, N.Y.; Liu, B.; Li, Z.Y. Highly Selective, Sensitive and Rapid Detection of Escherichia coli O157: H7 Using Duplex PCR and Magnetic Nanoparticle-Based Chemiluminescence Assay. J. Biomed. Nanotechnol. 2017, 13, 1243–1252. [Google Scholar] [CrossRef]
  31. Ling, Y.Z.; Zhu, Y.F.; Fan, H.H.; Zha, H.L.; Yang, M.; Wu, L.; Chen, H.; Li, W.; Wu, Y.P.; Chen, H.B. Rapid Method for Detection of Staphylococcus aureus in Feces. J. Biomed. Nanotechnol. 2019, 15, 1290–1298. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, H.N.; Dong, H.M.; Chen, Z.; Lin, L.; Chen, H.; Li, S.; Deng, Y. Magnetic Nanoparticles Enhanced Microarray Detection of Multiple Foodborne Pathogens. J. Biomed. Nanotechnol. 2017, 13, 1333–1343. [Google Scholar] [CrossRef]
  33. Li, S.; Liu, H.N.; Deng, Y.; Lin, L.; He, N.Y. Development of a Magnetic Nanoparticles Microarray for Simultaneous and Simple Detection of Foodborne Pathogens. J. Biomed. Nanotechnol. 2013, 9, 1254–1260. [Google Scholar] [CrossRef] [PubMed]
  34. Mou, X.B.; Chen, Z.; Li, T.T.; Liu, M.; Liu, Y.; Ali, Z.; Li, S.; Zhu, Y.B.; Li, Z.Y.; Deng, Y. A Highly Sensitive Strategy for Low-Abundance Hepatitis B Virus Detection via One-Step Nested Polymerase Chain Reaction, Chemiluminescence Technology and Magnetic Separation. J. Biomed. Nanotechnol. 2019, 15, 1832–1838. [Google Scholar] [CrossRef]
  35. Hao, H.Q.; Ma, Q.M.; He, F.; Yao, P. Doxorubicin and Fe3O4 loaded albumin nanoparticles with folic acid modified dextran surface for tumor diagnosis and therapy. J. Mater. Chem. B 2014, 2, 7978–7987. [Google Scholar] [CrossRef]
  36. Wang, Q.; Zhao, X.M.; Yan, H.; Kang, F.Y.; Li, Z.F.; Qiao, Y.Y.; Li, D. A cross-talk EGFR/VEGFR-targeted bispecific nanoprobe for magnetic resonance/near-infrared fluorescence imaging of colorectal cancer. MRS Commun. 2018, 8, 1008–1017. [Google Scholar] [CrossRef]
  37. Wang, M.L.; Yang, Q.M.; Li, M.; Zou, H.M.; Wang, Z.G.; Ran, H.T.; Zheng, Y.Y.; Jian, J.; Zhou, Y.; Luo, Y.D.; et al. Multifunctional Nanoparticles for Multimodal Imaging-Guided Low-Intensity Focused Ultrasound/Immunosynergistic Retinoblastoma Therapy. ACS Appl. Mater. Interfaces 2020, 12, 5642–5657. [Google Scholar] [CrossRef]
  38. Tang, Y.J.; Ali, Z.; Dai, J.G.; Liu, X.L.; Wu, Y.Q.; Chen, Z.; He, N.Y.; Li, S.; Wang, L.J. Single-Nucleotide Polymorphism Genotyping of exoS in Pseudomonas aeruginosa Using Dual-Color Fluorescence Hybridization and Magnetic Separation. J. Biomed. Nanotechnol. 2018, 14, 206–214. [Google Scholar] [CrossRef]
  39. Mou, X.B.; Sheng, D.N.; Chen, Z.; Liu, M.; Liu, Y.; Deng, Y.; Xu, K.; Hou, R.X.; Zhao, J.Y.; Zhu, Y.B.; et al. In-Situ Mutation Detection by Magnetic Beads-Probe Based on Single Base Extension and Its Application in Genotyping of Hepatitis B Virus Pre-C Region 1896nt Locus Single Nucleotide Polymorphisms. J. Biomed. Nanotechnol. 2019, 15, 2393–2400. [Google Scholar] [CrossRef]
  40. Liu, B.; Jia, Y.Y.; Ma, M.; Li, Z.Y.; Liu, H.N.; Li, S.; Deng, Y.; Zhang, L.M.; Lu, Z.X.; Wang, W.; et al. High Throughput SNP Detection System Based on Magnetic Nanoparticles Separation. J. Biomed. Nanotechnol. 2013, 9, 247–256. [Google Scholar] [CrossRef] [Green Version]
  41. Li, S.; Liu, H.N.; Jia, Y.Y.; Mou, X.B.; Deng, Y.; Lin, L.; Liu, B.; He, N.Y. An Automatic High-Throughput Single Nucleotide Polymorphism Genotyping Approach Based on Universal Tagged Arrays and Magnetic Nanoparticles. J. Biomed. Nanotechnol. 2013, 9, 689–698. [Google Scholar] [CrossRef] [Green Version]
  42. Tang, C.L.; He, Z.Y.; Liu, H.M.; Xu, Y.Y.; Huang, H.; Yang, G.J.; Xiao, Z.Q.; Li, S.; Liu, H.N.; Deng, Y.; et al. Application of magnetic nanoparticles in nucleic acid detection. J. Nanobiotechnol. 2020, 18, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wang, Y.Z.; Shi, Z.; Sun, Y.; Wu, X.Y.; Li, S.; Dong, S.B. Preparation of amphiphilic magnetic polyvinyl alcohol targeted drug carrier and drug delivery research. Des. Monomers Polym. 2020, 23, 197–206. [Google Scholar] [CrossRef] [PubMed]
  44. Shen, L.Z.; Li, B.; Qiao, Y.S. Fe3O4 Nanoparticles in Targeted Drug/Gene Delivery Systems. Materials 2018, 11, 324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Yew, Y.P.; Shameli, K.; Miyake, M.; Khairudin, N.B.B.A.; Mohamad, S.E.B.; Naiki, T.; Lee, K.X. Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles and biomedical applications in targeted anticancer drug delivery system: A review. Arab. J. Chem. 2020, 13, 2287–2308. [Google Scholar] [CrossRef]
  46. Hu, Y.; Mignani, S.; Majoral, J.P.; Shen, M.W.; Shi, X.Y. Construction of iron oxide nanoparticle-based hybrid platforms for tumor imaging and therapy. Chem. Soc. Rev. 2018, 47, 1874–1900. [Google Scholar] [CrossRef]
  47. Wang, K.; Xu, X.G.; Ma, Y.L.; Sheng, C.R.; Li, L.N.; Lu, L.Y.; Wang, J.; Wang, Y.N.; Jiang, Y. Fe3O4@Angelica sinensis polysaccharide nanoparticles as an ultralow-toxicity contrast agent for magnetic resonance imaging. Rare Met. 2021, 40, 2486–2493. [Google Scholar] [CrossRef]
  48. Li, J.J.; Wang, J.H.; Li, J.Y.; Yang, X.; Wan, J.L.; Zheng, C.S.; Du, Q.; Zhou, G.F.; Yang, X.L. Fabrication of Fe3O4@PVA microspheres by one-step electrospray for magnetic resonance imaging during transcatheter arterial embolization. Acta Biomater. 2021, 131, 532–543. [Google Scholar] [CrossRef]
  49. Wu, C.H.; Zhao, W.W.; Yu, J.; Li, S.J.; Lin, L.G.; Chen, X.P. Induction of ferroptosis and mitochondrial dysfunction by oxidative stress in PC12 cells. Sci. Rep. 2018, 8, 574. [Google Scholar] [CrossRef] [Green Version]
  50. Chen, Y.; Liu, Y.; Lan, T.; Qin, W.; Zhu, Y.T.; Qin, K.; Gao, J.J.; Wang, H.B.; Hou, X.M.; Chen, N.; et al. Quantitative Profiling of Protein Carbonylations in Ferroptosis by an Aniline-Derived Probe. J. Am. Chem. Soc. 2018, 140, 4712–4720. [Google Scholar] [CrossRef] [Green Version]
  51. Latunde-Dada, G.O. Ferroptosis: Role of lipid peroxidation, iron and ferritinophagy. Biochim. Biophys. Acta-Gen. Subj. 2017, 1861, 1893–1900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhu, L.Y.; Meng, D.N.; Wang, X.; Chen, X.R. Ferroptosis-Driven Nanotherapeutics to Reverse Drug Resistance in Tumor Microenvironment. ACS Appl. Bio Mater. 2022, 5, 2481–2506. [Google Scholar] [CrossRef]
  53. Dixon, S.J. Ferroptosis: Bug or feature? Immunol. Rev. 2017, 277, 150–157. [Google Scholar] [CrossRef] [PubMed]
  54. Jiang, M.Y.; Hu, R.L.; Yu, R.X.; Tang, Y.W.; Li, J.R. A narrative review of mechanisms of ferroptosis in cancer: New challenges and opportunities. Ann. Transl. Med. 2021, 9, 20. [Google Scholar] [CrossRef] [PubMed]
  55. Wu, J.M.; Ma, L.F.; Wang, J.Y.; Qiao, Y.X. Mechanism of Ferroptosis and Its Research Progress in Lung Cancer. Chin. J. Lung Cancer 2020, 23, 811–817. [Google Scholar]
  56. Gao, M.H.; Monian, P.; Pan, Q.H.; Zhang, W.; Xiang, J.; Jiang, X.J. Ferroptosis is an autophagic cell death process. Cell Res. 2016, 26, 1021–1032. [Google Scholar] [CrossRef] [Green Version]
  57. He, Y.J.; Liu, X.Y.; Xing, L.; Wan, X.; Chang, X.; Jiang, H.L. Fenton reaction-independent ferroptosis therapy via glutathione and iron redox couple sequentially triggered lipid peroxide generator. Biomaterials 2020, 241, 119911. [Google Scholar] [CrossRef]
  58. Liu, T.; Liu, W.L.; Zhang, M.K.; Yu, W.Y.; Gao, F.; Li, C.X.; Wang, S.B.; Feng, J.; Zhang, X.Z. Ferrous-Supply-Regeneration Nanoengineering for Cancer-Cell-Specific Ferroptosis in Combination with Imaging-Guided Photodynamic Therapy. ACS Nano 2018, 12, 12181–12192. [Google Scholar] [CrossRef]
  59. Bogdan, A.R.; Miyazawa, M.; Hashimoto, K.; Tsuji, Y. Regulators of Iron Homeostasis: New Players in Metabolism, Cell Death, and Disease. Trends Biochem. Sci. 2016, 41, 274–286. [Google Scholar] [CrossRef] [Green Version]
  60. Angeli, J.P.F.; Krysko, D.V.; Conrad, M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer 2019, 19, 405–414. [Google Scholar] [CrossRef]
  61. Imai, H.; Matsuoka, M.; Kumagai, T.; Sakamoto, T.; Koumura, T. Lipid Peroxidation-Dependent Cell Death Regulated by GPx4 and Ferroptosis. In Apoptotic and Non-Apoptotic Cell Death; Nagata, S., Nakano, H., Eds.; Springer: Cham, Switzerland, 2017; pp. 143–170. [Google Scholar]
  62. Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Grocin, A.G.; da Silva, T.N.X.; Panzilius, E.; Scheel, C.H.; et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 2019, 575, 693–698. [Google Scholar] [CrossRef] [PubMed]
  63. Bartolini, D.; Dallaglio, K.; Torquato, P.; Piroddi, M.; Galli, F. Nrf2-p62 autophagy pathway and its response to oxidative stress in hepatocellular carcinoma. Transl. Res. 2018, 193, 54–71. [Google Scholar] [CrossRef] [PubMed]
  64. Kang, R.; Kroemer, G.; Tang, D.L. The tumor suppressor protein p53 and the ferroptosis network. Free Radic. Biol. Med. 2019, 133, 162–168. [Google Scholar] [CrossRef]
  65. Chu, B.; Kon, N.; Chen, D.L.; Li, T.Y.; Liu, T.; Jiang, L.; Song, S.J.; Tavana, O.; Gu, W. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat. Cell Biol. 2019, 21, 579–591. [Google Scholar] [CrossRef]
  66. Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; IngoId, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 2017, 13, 91–98. [Google Scholar] [CrossRef]
  67. Shen, Z.; Song, J.; Yung, B.C.; Zhou, Z.; Wu, A.; Chen, X. Emerging Strategies of Cancer Therapy Based on Ferroptosis. Adv. Mater. 2018, 30, e1704007. [Google Scholar] [CrossRef]
  68. Lai, Y.X.; Deng, Y.; Yang, G.J.; Li, S.; Zhang, C.X.; Liu, X.Y. Molecular Imprinting Polymers Electrochemical Sensor Based on AuNPs/PTh Modified GCE for Highly Sensitive Detection of Carcinomaembryonic Antigen. J. Biomed. Nanotechnol. 2018, 14, 1688–1694. [Google Scholar] [CrossRef]
  69. Lai, Y.X.; Wang, L.J.; Liu, Y.; Yang, G.J.; Tang, C.L.; Deng, Y.; Li, S. Immunosensors Based on Nanomaterials for Detection of Tumor Markers. J. Biomed. Nanotechnol. 2018, 14, 44–65. [Google Scholar] [CrossRef]
  70. Su, W.; Ma, L.; Wu, S.H.; Li, W.; Tang, J.X.; Deng, J.; Liu, J.X. Effect of Surface Modification of Silver Nanoparticles on the Proliferation of Human Lung Squamous Cell Carcinoma (HTB182) and Bronchial Epithelial (HBE) Cells In Vitro. J. Biomed. Nanotechnol. 2017, 13, 1281–1291. [Google Scholar] [CrossRef]
  71. Wu, Y.Y.; Deng, P.H.; Tian, Y.L.; Ding, Z.Y.; Li, G.L.; Liu, J.; Zuberi, Z.; He, Q.G. Rapid recognition and determination of tryptophan by carbon nanotubes and molecularly imprinted polymer-modified glassy carbon electrode. Bioelectrochemistry 2020, 131, 107393. [Google Scholar] [CrossRef]
  72. Gong, L.; Zhao, L.; Tan, M.D.; Pan, T.; He, H.; Wang, Y.L.; He, X.L.; Li, W.J.; Tang, L.; Nie, L.B. Two-Photon Fluorescent Nanomaterials and Their Applications in Biomedicine. J. Biomed. Nanotechnol. 2021, 17, 509–528. [Google Scholar] [CrossRef] [PubMed]
  73. Huang, L.; Su, E.B.; Liu, Y.; He, N.Y.; Deng, Y.; Jin, L.; Chen, Z.; Li, S. A microfluidic device for accurate detection of hs-cTnI. Chin. Chem. Lett. 2021, 32, 1555–1558. [Google Scholar] [CrossRef]
  74. Xiao, X.Y.; Yang, H.C.; Jiang, P.F.; Chen, Z.; Ji, C.Y.; Nie, L.B. Multi-Functional Fe3O4@mSiO(2)-AuNCs Composite Nanoparticles Used as Drug Delivery System. J. Biomed. Nanotechnol. 2017, 13, 1292–1299. [Google Scholar] [CrossRef]
  75. Zhao, H.; Su, E.B.; Huang, L.; Zai, Y.F.; Liu, Y.; Chen, Z.; Li, S.; Jin, L.; Deng, Y.; He, N.Y. Washing-free chemiluminescence immunoassay for rapid detection of cardiac troponin I in whole blood samples. Chin. Chem. Lett. 2022, 33, 743–746. [Google Scholar] [CrossRef]
  76. Rezaei, S.J.T.; Malekzadeh, A.M.; Ramazani, A.; Niknejad, H. pH-Sensitive Magnetite Nanoparticles Modified with Hyperbranched Polymers and Folic Acid for Targeted Imaging and Therapy. Curr. Drug Deliv. 2019, 16, 839–848. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, S.Z.; Yu, X.J.; Qian, Y.N.; Chen, W.; Shen, J.L. Multifunctional magnetic iron oxide nanoparticles: An advanced platform for cancer theranostics. Theranostics 2020, 10, 6278–6309. [Google Scholar] [CrossRef]
  78. Guo, L.L.; Chen, H.; He, N.Y.; Deng, Y. Effects of surface modifications on the physicochemical properties of iron oxide nanoparticles and their performance as anticancer drug carriers. Chin. Chem. Lett. 2018, 29, 1829–1833. [Google Scholar] [CrossRef]
  79. Shah, M.A.A.; He, N.Y.; Li, Z.Y.; Ali, Z.S.; Zhang, L.M. Nanoparticles for DNA Vaccine Delivery. J. Biomed. Nanotechnol. 2014, 10, 2332–2349. [Google Scholar] [CrossRef]
  80. Song, J.; Lin, L.; Yang, Z.; Zhu, R.; Zhou, Z.; Li, Z.-W.; Wang, F.; Chen, J.; Yang, H.; Chen, X. Self-Assembled Responsive Bilayered Vesicles with Adjustable Oxidative Stress for Enhanced Cancer Imaging and Therapy. J. Am. Chem. Soc. 2019, 141, 8158–8170. [Google Scholar] [CrossRef]
  81. Tian, X.; Ruan, L.; Zhou, S.; Wu, L.; Cao, J.; Qi, X.; Zhang, X.; Shen, S. Appropriate Size of Fe3O4 Nanoparticles for Cancer Therapy by Ferroptosis. ACS Appl. Bio Mater. 2022, 5, 1692–1699. [Google Scholar] [CrossRef]
  82. Trujillo-Alonso, V.; Pratt, E.C.; Zong, H.L.; Lara-Martinez, A.; Kaittanis, C.; Rabie, M.O.; Longo, V.; Becker, M.W.; Roboz, G.J.; Grimm, J.; et al. FDA-approved ferumoxytol displays anti-leukaemia efficacy against cells with low ferroportin levels. Nat. Nanotechnol. 2019, 14, 616–622. [Google Scholar] [CrossRef] [PubMed]
  83. Gao, J.; Luo, T.; Wang, J. Gene interfered-ferroptosis therapy for cancers. Nat. Commun. 2021, 12, 5311. [Google Scholar] [CrossRef]
  84. Zanganeh, S.; Hutter, G.; Spitler, R.; Lenkov, O.; Mahmoudi, M.; Shaw, A.; Pajarinen, J.S.; Nejadnik, H.; Goodman, S.; Moseley, M.; et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat. Nanotechnol. 2016, 11, 986–994. [Google Scholar] [CrossRef]
  85. Zhou, L.Y.; Peng, Y.B.; Wang, Q.Q.; Lin, Q.L. An ESIPT-based two-photon fluorescent probe detection of hydrogen peroxide in live cells and tissues. J. Photochem. Photobiol. B-Biol. 2017, 167, 264–268. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, S.; Li, D.; Yuan, Y. Long-term moderate intensity exercise alleviates myocardial fibrosis in type 2 diabetic rats via inhibitions of oxidative stress and TGF-β1/Smad pathway. J. Physiol. Sci. JPS 2019, 69, 861–873. [Google Scholar] [CrossRef] [PubMed]
  87. Deng, Y.; He, N.Y.; Xu, L.J.; Li, X.L.; Li, S.; Li, Z.Y.; Liu, H.N. A Rapid Scavenger of the Lipid Peroxidation Product Malondialdehyde: New Perspective of Taurine. Adv. Sci. Lett. 2011, 4, 442–448. [Google Scholar] [CrossRef]
  88. Jiang, L.; Kon, N.; Li, T.Y.; Wang, S.J.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef] [Green Version]
  89. Xie, Y.C.; Zhu, S.; Song, X.X.; Sun, X.F.; Fan, Y.; Liu, J.B.; Zhong, M.; Yuan, H.; Zhang, L.; Billiar, T.R.; et al. The Tumor Suppressor p53 Limits Ferroptosis by Blocking DPP4 Activity. Cell Rep. 2017, 20, 1692–1704. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, Z.; Xia, X.; Lv, X.; Song, E.; Song, Y. Iron-bearing nanoparticles trigger human umbilical vein endothelial cells ferroptotic responses by promoting intracellular iron level. Environ. Pollut. 2021, 287, 117345. [Google Scholar] [CrossRef]
  91. Wu, C.; Shen, Z.; Lu, Y.; Sun, F.; Shi, H. p53 Promotes Ferroptosis in Macrophages Treated with Fe3O4 Nanoparticles. ACS Appl. Mater. Interfaces 2022, 14, 42791–42803. [Google Scholar] [CrossRef]
  92. Zhang, J.; Kong, X.; Zhang, Y.; Sun, W.; Xu, E.; Chen, X. Mdm2 is a target and mediator of IRP2 in cell growth control. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2020, 34, 2301–2311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhou, Y.; Que, K.; Zhang, Z.; Yi, Z.; Zhao, P.; You, Y.; Gong, J.; Liu, Z. Iron overloaded polarizes macrophage to proinflammation phenotype through ROS/acetyl-p53 pathway. Cancer Med. 2018, 7, 4012–4022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhang, Y.L.; Koppula, P.; Gan, B.Y. Regulation of H2A ubiquitination and SLC7A11 expression by BAP1 and PRC1. Cell Cycle 2019, 18, 773–783. [Google Scholar] [CrossRef] [Green Version]
  95. Wen, J.; Chen, H.; Ren, Z.; Zhang, P.; Chen, J.; Jiang, S. Ultrasmall iron oxide nanoparticles induced ferroptosis via Beclin1/ATG5-dependent autophagy pathway. Nano Converg. 2021, 8, 10. [Google Scholar] [CrossRef]
  96. Chen, X.; Kang, R.; Kroemer, G.; Tang, D.L. Broadening horizons: The role of ferroptosis in cancer. Nat. Rev. Clin. Oncol. 2021, 18, 280–296. [Google Scholar] [CrossRef]
  97. Yu, H.T.; Guo, P.Y.; Xie, X.Z.; Wang, Y.; Chen, G. Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J. Cell. Mol. Med. 2017, 21, 648–657. [Google Scholar] [CrossRef] [Green Version]
  98. Wu, Y.N.; Yu, C.C.; Luo, M.; Cen, C.; Qiu, J.L.; Zhang, S.Z.; Hu, K.M. Ferroptosis in Cancer Treatment: Another Way to Rome. Front. Oncol. 2020, 10, 571127. [Google Scholar] [CrossRef]
  99. Yang, G.J.; Lai, Y.X.; Xiao, Z.Q.; Tang, C.L.; Deng, Y. Ultrasensitive electrochemical immunosensor of carcinoembryonic antigen based on gold-label silver-stain signal amplification. Chin. Chem. Lett. 2018, 29, 1857–1860. [Google Scholar] [CrossRef]
  100. Mou, X.B.; Ali, Z.; Li, B.; Li, T.T.; Yi, H.; Dong, H.M.; He, N.Y.; Deng, Y.; Zeng, X. Multiple genotyping based on multiplex PCR and microarray. Chin. Chem. Lett. 2016, 27, 1661–1665. [Google Scholar] [CrossRef]
  101. Wu, Y.Y.; Deng, P.H.; Tian, Y.L.; Feng, J.X.; Xiao, J.Y.; Li, J.H.; Liu, J.; Li, G.L.; He, Q.G. Simultaneous and sensitive determination of ascorbic acid, dopamine and uric acid via an electrochemical sensor based on PVP-graphene composite. J. Nanobiotechnol. 2020, 18, 112. [Google Scholar] [CrossRef]
  102. Gao, Z.; He, T.; Zhang, P.; Li, X.; Zhang, Y.; Lin, J.; Hao, J.; Huang, P.; Cui, J. Polypeptide-Based Theranostics with Tumor-Microenvironment-Activatable Cascade Reaction for Chemo-ferroptosis Combination Therapy. ACS Appl. Mater. Interfaces 2020, 12, 20271–20280. [Google Scholar] [CrossRef] [PubMed]
  103. Guo, J.P.; Xu, B.F.; Han, Q.; Zhou, H.X.; Xia, Y.; Gong, C.W.; Dai, X.F.; Li, Z.Y.; Wu, G. Ferroptosis: A Novel Anti-tumor Action for Cisplatin. Cancer Res. Treat. 2018, 50, 445–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Liu, B.; Ji, Q.; Cheng, Y.; Liu, M.; Zhang, B.; Mei, Q.; Liu, D.; Zhou, S. Biomimetic GBM-targeted drug delivery system boosting ferroptosis for immunotherapy of orthotopic drug-resistant GBM. J. Nanobiotechnol. 2022, 20, 161. [Google Scholar] [CrossRef] [PubMed]
  105. Cloughesy, T.; Mochizuki, A.; Orpilla, J.; Hugo, W.; Lee, A.; Davidson, T.; Wang, A.; Ellingson, B.; Rytlewski, J.; Sanders, C.; et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019, 25, 477–486. [Google Scholar] [CrossRef]
  106. Chen, M.; Li, J.; Shu, G.; Shen, L.; Qiao, E.; Zhang, N.; Fang, S.; Chen, X.; Zhao, Z.; Tu, J.; et al. Homogenous multifunctional microspheres induce ferroptosis to promote the anti-hepatocarcinoma effect of chemoembolization. J. Nanobiotechnol. 2022, 20, 179. [Google Scholar] [CrossRef]
  107. Wilhelm, S.; Tavares, A.J.; Dai, Q.; Ohta, S.; Audet, J.; Dvorak, H.F.; Chan, W.C.W. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 2016, 1, 16014. [Google Scholar] [CrossRef]
  108. Yao, X.; Xie, R.; Cao, Y.; Tang, J.; Men, Y.; Peng, H.; Yang, W. Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J. Nanobiotechnol. 2021, 19, 311. [Google Scholar] [CrossRef]
  109. Wu, F.; Du, Y.; Yang, J.; Shao, B.; Mi, Z.; Yao, Y.; Cui, Y.; He, F.; Zhang, Y.; Yang, P. Peroxidase-like Active Nanomedicine with Dual Glutathione Depletion Property to Restore Oxaliplatin Chemosensitivity and Promote Programmed Cell Death. ACS Nano 2022, 16, 3647–3663. [Google Scholar] [CrossRef]
  110. Wang, X.; Li, P.; Jing, X.; Zhou, Y.; Shao, Y.; Zheng, M.; Wang, J.; Ran, H.; Tang, H. Folate-modified erythrocyte membrane nanoparticles loaded with Fe3O4 and artemisinin enhance ferroptosis of tumors by low-intensity focused ultrasound. Front. Oncol. 2022, 12, 864444. [Google Scholar] [CrossRef]
  111. Yang, H.W.; Liang, W.B.; Si, J.; Li, Z.Y.; He, N.Y. Long Spacer Arm-Functionalized Magnetic Nanoparticle Platform for Enhanced Chemiluminescent Detection of Hepatitis B Virus. J. Biomed. Nanotechnol. 2014, 10, 3610–3619. [Google Scholar] [CrossRef]
  112. Fang, Y.L.; Liu, H.R.; Wang, Y.; Su, X.Y.; Jin, L.; Wu, Y.Q.; Deng, Y.; Li, S.; Chen, Z.; Chen, H.; et al. Fast and Accurate Control Strategy for Portable Nucleic Acid Detection (PNAD) System Based on Magnetic Nanoparticles. J. Biomed. Nanotechnol. 2021, 17, 407–415. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, H.; Wu, Y.Q.; Chen, Z.; Hu, Z.L.; Fang, Y.L.; Liao, P.; Deng, Y.; He, N.Y. Performance Evaluation of a Novel Sample In-Answer Out (SIAO) System Based on Magnetic Nanoparticles. J. Biomed. Nanotechnol. 2017, 13, 1619–1630. [Google Scholar] [CrossRef] [PubMed]
  114. Ma, C.; Li, C.Y.; Wang, F.; Ma, N.N.; Li, X.L.; Li, Z.Y.; Deng, Y.; Wang, Z.F.; Xi, Z.J.; Tang, Y.J.; et al. Magnetic Nanoparticles-Based Extraction and Verification of Nucleic Acids from Different Sources. J. Biomed. Nanotechnol. 2013, 9, 703–709. [Google Scholar] [CrossRef] [PubMed]
  115. Yang, H.W.; Liu, M.; Jiang, H.R.; Zeng, Y.; Jin, L.; Luan, T.; Deng, Y.; He, N.Y.; Zhang, G.; Zeng, X. Copy Number Variation Analysis Based on Gold Magnetic Nanoparticles and Fluorescence Multiplex Ligation-Dependent Probe Amplification. J. Biomed. Nanotechnol. 2017, 13, 655–664. [Google Scholar] [CrossRef]
  116. Guo, L.L.; Wang, T.; Chen, Z.; He, N.Y.; Chen, Y.Z.; Yuan, T. Light scattering based analyses of the effects of bovine serum proteins on interactions of magnetite spherical particles with cells. Chin. Chem. Lett. 2018, 29, 1291–1295. [Google Scholar] [CrossRef]
  117. Liang, H.; Wu, X.; Zhao, G.; Feng, K.; Ni, K.; Sun, X. Renal Clearable Ultrasmall Single-Crystal Fe Nanoparticles for Highly Selective and Effective Ferroptosis Therapy and Immunotherapy. J. Am. Chem. Soc. 2021, 143, 15812–15823. [Google Scholar] [CrossRef]
  118. Zhang, Y.; Wang, F.M.; Liu, C.Q.; Wang, Z.Z.; Kang, L.H.; Huang, Y.Y.; Dong, K.; Ren, J.S.; Qu, X.G. Nanozyme Decorated Metal-Organic Frameworks for Enhanced Photodynamic Therapy. ACS Nano 2018, 12, 651–661. [Google Scholar] [CrossRef]
  119. Shen, Z.Y.; Liu, T.; Li, Y.; Lau, J.; Yang, Z.; Fan, W.P.; Zhou, Z.J.; Shi, C.R.; Ke, C.M.; Bregadze, V.I.; et al. Fenton-Reaction-Acceleratable Magnetic Nanoparticles for Ferroptosis Therapy of Orthotopic Brain Tumors. ACS Nano 2018, 12, 11355–11365. [Google Scholar] [CrossRef]
  120. Tang, Z.; Liu, Y.; He, M.; Bu, W. Chemodynamic Therapy: Tumour Microenvironment-Mediated Fenton and Fenton-like Reactions. Angew. Chem. Int. Ed. Engl. 2019, 58, 946–956. [Google Scholar] [CrossRef]
  121. Chen, Q.; Ma, X.; Xie, L.; Chen, W.; Xu, Z.; Song, E.; Zhu, X.; Song, Y. Iron-based nanoparticles for MR imaging-guided ferroptosis in combination with photodynamic therapy to enhance cancer treatment. Nanoscale 2021, 13, 4855–4870. [Google Scholar] [CrossRef]
  122. Liang, X.; Chen, M.; Bhattarai, P.; Hameed, S.; Tang, Y.; Dai, Z. Complementing Cancer Photodynamic Therapy with Ferroptosis through Iron Oxide Loaded Porphyrin-Grafted Lipid Nanoparticles. ACS Nano 2021, 15, 20164–20180. [Google Scholar] [CrossRef] [PubMed]
  123. Jiang, Y.Y.; Zhao, X.H.; Huang, J.G.; Li, J.C.; Upputuri, P.K.; Sun, H.; Han, X.; Pramanik, M.; Miao, Y.S.; Duan, H.W.; et al. Transformable hybrid semiconducting polymer nanozyme for second near-infrared photothermal ferrotherapy. Nat. Commun. 2020, 11, 1857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef] [Green Version]
  125. Xie, S.; Sun, W.; Zhang, C.; Dong, B.; Yang, J.; Hou, M.; Xiong, L.; Cai, B.; Liu, X.; Xue, W. Metabolic Control by Heat Stress Determining Cell Fate to Ferroptosis for Effective Cancer Therapy. ACS Nano 2021, 15, 7179–7194. [Google Scholar] [CrossRef]
  126. Qian, X.; Zhang, J.; Gu, Z.; Chen, Y. Nanocatalysts-augmented Fenton chemical reaction for nanocatalytic tumor therapy. Biomaterials 2019, 211, 1–13. [Google Scholar] [CrossRef] [PubMed]
  127. Wu, F.X.; Chen, H.R.; Liu, R.Q.; Suo, Y.K.; Li, Q.Q.; Zhang, Y.L.; Liu, H.G.; Cheng, Z.; Chang, Y.L. An active-passive strategy for enhanced synergistic photothermal-ferroptosis therapy in the NIR-I/II biowindows. Biomater. Sci. 2022, 10, 1104–1112. [Google Scholar] [CrossRef] [PubMed]
  128. Antoniak, M.A.; Pazik, R.; Bazylinska, U.; Wiwatowski, K.; Tomaszewska, A.; Kulpa-Greszta, M.; Adamczyk-Grochala, J.; Wnuk, M.; Mackowski, S.; Lewinska, A.; et al. Multimodal polymer encapsulated CdSe/Fe3O4 nanoplatform with improved biocompatibility for two-photon and temperature stimulated bioapplications. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 127, 112224. [Google Scholar] [CrossRef]
  129. Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12, 991–1003. [Google Scholar] [CrossRef]
  130. Li, B.B.; Xu, Q.N.; Li, X.F.; Zhang, P.; Zhao, X.; Wang, Y.X. Redox-responsive hyaluronic acid nanogels for hyperthermia-assisted chemotherapy to overcome multidrug resistance. Carbohydr. Polym. 2019, 203, 378–385. [Google Scholar] [CrossRef]
  131. Chen, M.J.; Zhang, F.; Song, J.J.; Weng, Q.Y.; Li, P.C.; Li, Q.; Qian, K.; Ji, H.X.; Pietrini, S.; Ji, J.S.; et al. Image-Guided Peri-Tumoral Radiofrequency Hyperthermia-Enhanced Direct Chemo-Destruction of Hepatic Tumor Margins. Front. Oncol. 2021, 11, 593996. [Google Scholar] [CrossRef]
  132. Zhu, M.T.; Wu, P.Y.; Li, Y.; Zhang, L.; Zong, Y.J.; Wan, M.X. Synergistic therapy for orthotopic gliomas via biomimetic nanosonosensitizer-mediated sonodynamic therapy and ferroptosis. Biomater. Sci. 2022, 10, 3911–3923. [Google Scholar] [CrossRef] [PubMed]
  133. Liang, C.; Zhang, X.L.; Yang, M.S.; Dong, X.C. Recent Progress in Ferroptosis Inducers for Cancer Therapy. Adv. Mater. 2019, 31, 1904197. [Google Scholar] [CrossRef] [PubMed]
  134. Galadari, S.; Rahman, A.; Pallichankandy, S.; Thayyullathil, F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic. Biol. Med. 2017, 104, 144–164. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Major metabolic pathways of ferroptosis. By Figdraw.
Figure 1. Major metabolic pathways of ferroptosis. By Figdraw.
Molecules 28 04562 g001
Figure 2. Effect of Fe3O4-NPs on cell ferroptosis. By Figdraw.
Figure 2. Effect of Fe3O4-NPs on cell ferroptosis. By Figdraw.
Molecules 28 04562 g002
Figure 3. Effect of particle size parameters of Fe3O4-NPs on cell ferroptosis. By Figdraw.
Figure 3. Effect of particle size parameters of Fe3O4-NPs on cell ferroptosis. By Figdraw.
Molecules 28 04562 g003
Figure 4. Mechanism of Fe3O4-NP-induced ferroptosis through up-regulation of p53. By Figdraw.
Figure 4. Mechanism of Fe3O4-NP-induced ferroptosis through up-regulation of p53. By Figdraw.
Molecules 28 04562 g004
Figure 5. Mechanism of Pt&Fe3O4@PP-induced ferroptosis. By Figdraw.
Figure 5. Mechanism of Pt&Fe3O4@PP-induced ferroptosis. By Figdraw.
Molecules 28 04562 g005
Figure 6. Mechanism of DFMC-induced ferroptosis. By Figdraw.
Figure 6. Mechanism of DFMC-induced ferroptosis. By Figdraw.
Molecules 28 04562 g006
Figure 7. Mechanism of PLGA-induced ferroptosis. By Figdraw.
Figure 7. Mechanism of PLGA-induced ferroptosis. By Figdraw.
Molecules 28 04562 g007
Figure 8. Mechanism of GBP@Fe3O4-induced ferroptosis. By Figdraw.
Figure 8. Mechanism of GBP@Fe3O4-induced ferroptosis. By Figdraw.
Molecules 28 04562 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, Y.; Wu, X.; Bao, X.; Mou, X. Progress in the Mechanism of the Effect of Fe3O4 Nanomaterials on Ferroptosis in Tumor Cells. Molecules 2023, 28, 4562. https://doi.org/10.3390/molecules28114562

AMA Style

Wang Y, Wu X, Bao X, Mou X. Progress in the Mechanism of the Effect of Fe3O4 Nanomaterials on Ferroptosis in Tumor Cells. Molecules. 2023; 28(11):4562. https://doi.org/10.3390/molecules28114562

Chicago/Turabian Style

Wang, Yaxuan, Xiao Wu, Xiaoying Bao, and Xianbo Mou. 2023. "Progress in the Mechanism of the Effect of Fe3O4 Nanomaterials on Ferroptosis in Tumor Cells" Molecules 28, no. 11: 4562. https://doi.org/10.3390/molecules28114562

Article Metrics

Back to TopTop