Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 468: 115028, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38723677

ABSTRACT

Early life stress (ELS) increases the risk of depression later in life. Programmed cell death factor 4 (PDCD4), an apoptosis-related molecule, extensively participates in tumorigenesis and inflammatory diseases. However, its involvement in a person's susceptibility to ELS-related depression is unknown. To examine the effects and underlying mechanisms of PDCD4 on ELS vulnerability, we used a "two-hit" stress mouse model: an intraperitoneal injection of lipopolysaccharide (LPS) into neonatal mice was performed on postnatal days 7-9 (P7-P9) and inescapable foot shock (IFS) administration in adolescent was used as a later-life challenge. Our study shows that compared with mice that were only exposed to the LPS or IFS, the "two-hit" stress mice developed more severe depression/anxiety-like behaviors and social disability. We detected the levels of PDCD4 in the hippocampus of adolescent mice and found that they were significantly increased in "two-hit" stress mice. The results of immunohistochemical staining and Sholl analysis showed that the number of microglia in the hippocampus of "two-hit" stress mice significantly increased, with morphological changes, shortened branches, and decreased numbers. However, knocking down PDCD4 can prevent the number and morphological changes of microglia induced by ELS. In addition, we confirmed through the Golgi staining and immunohistochemical staining results that knocking down PDCD4 can ameliorate ELS-induced synaptic plasticity damage. Mechanically, the knockdown of PDCD4 exerts neuroprotective effects, possibly via the mediation of BDNF/AKT/CREB signaling. Combined, these results suggest that PDCD4 may play an important role in the ELS-induced susceptibility to depression and, thus, may become a therapeutic target for depressive disorders.


Subject(s)
Apoptosis Regulatory Proteins , Depression , Hippocampus , Mice, Inbred C57BL , Neuronal Plasticity , RNA-Binding Proteins , Stress, Psychological , Animals , Male , Mice , Animals, Newborn , Apoptosis Regulatory Proteins/metabolism , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/metabolism , Depression/metabolism , Depression/physiopathology , Disease Models, Animal , Disease Susceptibility , Hippocampus/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism , Neuronal Plasticity/physiology , RNA-Binding Proteins/metabolism , Stress, Psychological/metabolism , Female
2.
J Control Release ; 365: 1074-1088, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38101752

ABSTRACT

Oxidative stress is a major obstacle for neurological functional recovery after hypoxia-ischemia (HI) brain damage. Nanozymes with robust anti-oxidative stress properties offer a therapeutic option for HI injury. However, insufficiency of nanozyme accumulation in the HI brain by noninvasive administration hinders their application. Herein, we reported a cerium vanadate (CeVO4) nanozyme to realize a noninvasive therapy for HI brain in neonatal mice by targeting brain neuron mitochondria. CeVO4 nanozyme with superoxide dismutase activity mainly co-located with neuronal mitochondria 1 h after administration. Pre- and post-HI administrations of CeVO4 nanozyme were able to attenuate acute brain injury, by inhibiting caspase-3 activation, microglia activation, and proinflammation cytokine production in the lesioned cortex 2 d after HI injury. Moreover, CeVO4 nanozyme administration led to short- and long-term functional recovery following HI insult without any potential toxicities in peripheral organs of mice even after prolonged delivery for 4 weeks. These beneficial effects of CeVO4 nanozyme were associated with suppressed oxidative stress and up-regulated nuclear factor erythroid-2-related factor 2 (Nrf2) expression. Finally, we found that Nrf2 inhibition with ML385 abolished the protective effects of CeVO4 nanozyme on HI injury. Collectively, this strategy may provide an applicative perspective for CeVO4 nanozyme therapy in HI brain damage via noninvasive delivery.


Subject(s)
Hypoxia-Ischemia, Brain , Vanadates , Animals , Mice , Animals, Newborn , Vanadates/therapeutic use , Vanadates/metabolism , Vanadates/pharmacology , Administration, Intranasal , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/pharmacology , NF-E2-Related Factor 2/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Brain/metabolism , Ischemia/drug therapy , Mitochondria
3.
Neural Regen Res ; 19(5): 1084-1094, 2024 May.
Article in English | MEDLINE | ID: mdl-37862212

ABSTRACT

We previously showed that hydrogen sulfide (H2S) has a neuroprotective effect in the context of hypoxic ischemic brain injury in neonatal mice. However, the precise mechanism underlying the role of H2S in this situation remains unclear. In this study, we used a neonatal mouse model of hypoxic ischemic brain injury and a lipopolysaccharide-stimulated BV2 cell model and found that treatment with L-cysteine, a H2S precursor, attenuated the cerebral infarction and cerebral atrophy induced by hypoxia and ischemia and increased the expression of miR-9-5p and cystathionine ß synthase (a major H2S synthetase in the brain) in the prefrontal cortex. We also found that an miR-9-5p inhibitor blocked the expression of cystathionine ß synthase in the prefrontal cortex in mice with brain injury caused by hypoxia and ischemia. Furthermore, miR-9-5p overexpression increased cystathionine-ß-synthase and H2S expression in the injured prefrontal cortex of mice with hypoxic ischemic brain injury. L-cysteine decreased the expression of CXCL11, an miR-9-5p target gene, in the prefrontal cortex of the mouse model and in lipopolysaccharide-stimulated BV-2 cells and increased the levels of proinflammatory cytokines BNIP3, FSTL1, SOCS2 and SOCS5, while treatment with an miR-9-5p inhibitor reversed these changes. These findings suggest that H2S can reduce neuroinflammation in a neonatal mouse model of hypoxic ischemic brain injury through regulating the miR-9-5p/CXCL11 axis and restoring ß-synthase expression, thereby playing a role in reducing neuroinflammation in hypoxic ischemic brain injury.

4.
Drug Des Devel Ther ; 17: 1175-1189, 2023.
Article in English | MEDLINE | ID: mdl-37113470

ABSTRACT

Introduction: Hypoxia-ischemia (HI) remains the leading cause of cerebral palsy and long-term neurological sequelae in infants. Despite intensive research and many therapeutic approaches, there are limited neuroprotective strategies against HI insults. Herein, we reported that HI insult significantly down-regulated microRNA-9-5p (miR-9-5p) level in the ipsilateral cortex of neonatal mice. Methods: The biological function and expression patterns of protein in the ischemic hemispheres were evaluated by qRT-PCR, Western Blotting analysis, Immunofluorescence and Immunohistochemistry. Open field test and Y-maze test were applied to detect locomotor activity and exploratory behavior and working memory. Results: Overexpression of miR-9-5p effectively alleviated brain injury and improved neurological behaviors following HI insult, accompanying with suppressed neuroinflammation and apoptosis. MiR-9-5p directly bound to the 3' untranslated region of DNA damage-inducible transcript 4 (DDIT4) and negatively regulated its expression. Furthermore, miR-9-5p mimics treatment down-regulated light chain 3 II/light chain 3 I (LC3 II/LC3 I) ratio and Beclin-1 expression and decreased LC3B accumulation in the ipsilateral cortex. Further analysis showed that DDIT4 knockdown conspicuously inhibited the HI-up-regulated LC3 II/ LC3 I ratio and Beclin-1 expression, associating with attenuated brain damage. Conclusion: The study indicates that miR-9-5p-mediated HI injury is regulated by DDIT4-mediated autophagy pathway and up-regulation of miR-9-5p level may provide a potential therapeutic effect on HI brain damage.


Subject(s)
Hypoxia-Ischemia, Brain , MicroRNAs , Animals , Mice , Animals, Newborn , Up-Regulation , Beclin-1/genetics , MicroRNAs/metabolism , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia/metabolism , Ischemia/metabolism , Brain/metabolism , Autophagy/physiology , Apoptosis , Transcription Factors/metabolism
5.
Cells ; 12(6)2023 03 09.
Article in English | MEDLINE | ID: mdl-36980197

ABSTRACT

We previously found that osteopontin (OPN) played a role in hypoxia-ischemia (HI) brain damage. However, its underlying mechanism is still unknown. Bioinformatics analysis revealed that the OPN protein was linked to the lysosomal cathepsin B (CTSB) and galectin-3 (GAL-3) proteins after HI exposure. In the present study, we tested the hypothesis that OPN was able to play a critical role in the lysosomal damage of microglia/macrophages following HI insult in neonatal mice. The results showed that OPN expression was enhanced, especially in microglia/macrophages, and colocalized with lysosomal-associated membrane protein 1 (LAMP1) and GAL-3; this was accompanied by increased LAMP1 and GAL-3 expression, CTSB leakage, as well as impairment of autophagic flux in the early stage of the HI process. In addition, the knockdown of OPN expression markedly restored lysosomal function with significant improvements in the autophagic flux after HI insult. Interestingly, cleavage of OPN was observed in the ipsilateral cortex following HI. The wild-type OPN and C-terminal OPN (Leu152-Asn294), rather than N-terminal OPN (Met1-Gly151), interacted with GAL-3 to induce lysosomal damage. Furthermore, the secreted OPN stimulated lysosomal damage by binding to CD44 in microglia in vitro. Collectively, this study demonstrated that upregulated OPN in microglia/macrophages and its cleavage product was able to interact with GAL-3, and secreted OPN combined with CD44, leading to lysosomal damage and exacerbating autophagosome accumulation after HI exposure.


Subject(s)
Brain Injuries , Hypoxia-Ischemia, Brain , Animals , Mice , Microglia/metabolism , Osteopontin/metabolism , Macrophages/metabolism , Brain Injuries/metabolism , Hypoxia-Ischemia, Brain/metabolism , Lysosomes/metabolism , Brain/metabolism
6.
J Affect Disord ; 330: 83-93, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36842657

ABSTRACT

INTRODUCTION: Neuroinflammation plays an important role in the development of major depressive disorder (MDD). Osteopontin (OPN) is one of the key molecules involved in neuroinflammation. We demonstrate here for the first time a key role of OPN in lipopolysaccharide (LPS)-induced depressive-like behavioral syndrome. METHODS: Systemic administration of LPS (5 mg/kg) mimics distinct depressive-like behavior, which could significantly upregulate OPN expression in microglia/macrophage in the hippocampus. The neurobehavioral assessments, quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR), Western blot, immunofluorescent staining, flow cytometry cell staining and Golgi staining were performed. RESULTS: Similar to fluoxetine treatment (the positive control), OPN knockdown with shRNA lentivirus markedly reversed LPS-induced depressive-like behavior. Moreover, knockdown of OPN suppressed LPS-induced proinflammatory cytokine expression, microglial activation, dendritic spines loss, as well as unregulated PSD-95 and BDNF in the hippocampus. CONCLUSION: We demonstrated that targeting OPN expression in microglia/macrophage might help to rescue LPS-induced depressive-like behavior. The underlying mechanism may relate to the modulation of neuroinflammation, BDNF signaling and synaptic structural complexity.


Subject(s)
Depressive Disorder, Major , Lipopolysaccharides , Mice , Animals , Lipopolysaccharides/pharmacology , Osteopontin/genetics , Neuroinflammatory Diseases , Brain-Derived Neurotrophic Factor
7.
Adv Sci (Weinh) ; 10(3): e2204528, 2023 01.
Article in English | MEDLINE | ID: mdl-36453595

ABSTRACT

Spinal cord injury (SCI) damages signal connections and conductions, with the result that neuronal circuits are disrupted leading to neural dysfunctions. Such injuries represent a serious and relatively common central nervous system condition and current treatments have limited success in the reconstruction of nerve connections in injured areas, especially where sizeable gaps are present. Biomaterial scaffolds have become an effective alternative to nerve transplantation in filling these gaps and provide the foundation for simulating the 3D structure of solid organs. However, there remain some limitations with the application of 3D bioprinting for preparation of biomaterial scaffolds. Here, the approach in constructing and testing mini-tissue building blocks and self-assembly, solid 3D gelatin microsphere (GM) scaffolds with multiple voids as based on the convenient preparation of gelatin microspheres by microfluidic devices is described. These 3D GM scaffolds demonstrate suitable biocompatibility, biodegradation, porosity, low preparation costs, and relative ease of production. Moreover, 3D GM scaffolds can effectively bridge injury gaps, establish nerve connections and signal transductions, mitigate inflammatory microenvironments, and reduce glial scar formation. Accordingly, these 3D GM scaffolds can serve as a novel and effective bridging method to promote nerve regeneration and reconstruction and thus recovery of nerve function after SCI.


Subject(s)
Gelatin , Spinal Cord Injuries , Rats , Animals , Microspheres , Tissue Scaffolds/chemistry , Spinal Cord Injuries/therapy , Biocompatible Materials
8.
Neural Regen Res ; 17(10): 2238-2246, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35259844

ABSTRACT

Extracellular vesicles (EVs) from mesenchymal stromal cells (MSCs) have previously been shown to protect against brain injury caused by hypoxia-ischemia (HI). The neuroprotective effects have been found to relate to the anti-inflammatory effects of EVs. However, the underlying mechanisms have not previously been determined. In this study, we induced oxygen-glucose deprivation in BV-2 cells (a microglia cell line), which mimics HI in vitro, and found that treatment with MSCs-EVs increased the cell viability. The treatment was also found to reduce the expression of pro-inflammatory cytokines, induce the polarization of microglia towards the M2 phenotype, and suppress the phosphorylation of selective signal transducer and activator of transcription 3 (STAT3) in the microglia. These results were also obtained in vivo using neonatal mice with induced HI. We investigated the potential role of miR-21a-5p in mediating these effects, as it is the most highly expressed miRNA in MSCs-EVs and interacts with the STAT3 pathway. We found that treatment with MSCs-EVs increased the levels of miR-21a-5p in BV-2 cells, which had been lowered following oxygen-glucose deprivation. When the level of miR-21a-5p in the MSCs-EVs was reduced, the effects on microglial polarization and STAT3 phosphorylation were reduced, for both the in vitro and in vivo HI models. These results indicate that MSCs-EVs attenuate HI brain injury in neonatal mice by shuttling miR-21a-5p, which induces microglial M2 polarization by targeting STAT3.

9.
Nanoscale Res Lett ; 16(1): 37, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33620584

ABSTRACT

Suppression of tumor development by inducing ferroptosis may provide a potential remedy for triple-negative breast cancer, which is sensitive to intracellular oxidative imbalance. Recently, artemisinin (ART) and its derivatives have been investigated as potential anticancer agents for the treatment of highly aggressive cancers via the induction of ferroptosis by iron-mediated cleavage of the endoperoxide bridge. Owing to its poor water solubility and limited intracellular iron content, it is challenging for further application in antitumor therapy. Herein, we developed ferrous-supply nano-carrier for ART based on tannic acid (TA) and ferrous ion (Fe(II)) coated on the zeolitic imidazolate framework-8 (ZIF) with ART encapsulated (TA-Fe/ART@ZIF) via coordination-driven self-assembly. Drug release experiments showed that ART was not nearly released in pH 7.4, while 59% ART was released in pH 5.0 after 10 h, demonstrating the excellent pH-triggered release. Meanwhile, a high level of intracellular ROS and MDA, accompanied with decreasing GSH and GPX4, displayed a newly developed nano-drug system displayed markedly enhanced ferroptosis. Compared with monotherapy, in vitro and vivo tumor inhibition experiments demonstrated higher efficiency of tumor suppression of TA-Fe/ART@ZIF. This work provides a novel approach to enhance the potency of ferroptotic nano-medicine and new directions for TBNC therapy.

10.
Mater Sci Eng C Mater Biol Appl ; 119: 111442, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33321583

ABSTRACT

Chemotherapy plays crucial roles in the clinical treatment of non-small cell lung cancer (NSCLC). Nevertheless, acquired chemoresistance is a common and critical problem that limits the clinical application of chemotherapy. Quercetin (QUE), a natural bioflavonoid, has significant antitumor potential, which has been verified in many drug-resistant cancer cell lines and animal models. Here, we explored whether QUE could reverse the resistance of NSCLC to paclitaxel (PTX)-based therapy. The results of cell viability revealed that QUE could synergistically enhance the cytotoxicity of PTX in A549 and A549/Taxol cells. Furthermore, Akt and ERK phosphorylation had no significant changes in A549/Taxol cells treated with PTX. However, it was significantly inhibited by the combination treatment of QUE and PTX. To improve the antitumor activity of PTX due to its hydrophobicity and eliminate its toxicity, we prepared targeted biodegradable cetuximab chitosan nanoparticles (Cet-CTS NPs) to deliver PTX and QUE using ionic cross-linking technique. The targeted NPs displayed a particle size of 290 nm and sustained release of PTX and QUE. In addition, the targeted Cet-CTS NPs loaded with PTX and QUE inhibited tumor growth in PTX-resistant A549/Taxol cells. Cet-QUE NPs decreased tumor growth in PTX-resistant xenografts. In conclusion, the administration of QUE by using Cet-CTS NPs could provide a prospective strategy for the treatment of PTX-resistant lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Chitosan , Lung Neoplasms , Nanoparticles , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Lung Neoplasms/drug therapy , Paclitaxel/pharmacology , Prospective Studies , Quercetin/pharmacology
11.
Cell Death Dis ; 11(9): 751, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929075

ABSTRACT

Although ferroptosis has been recognized as a novel antitumoral treatment, high expression of nuclear factor erythroid 2-related factor 2 (NRF2) has been reported to be an antioxidant transcript factor that protects malignant cells from ferroptosis. Previous findings indicated that metallothionein 1D pseudogene (MT1DP), a long noncoding RNA (lncRNA), functioned to aggravate oxidative stress by repressing antioxidation. Here we aimed at assessing whether MT1DP could regulate erastin-induced ferroptosis on non-small cell lung cancer (NSCLC) and elucidating the mechanism. We found that ectopic expression of MT1DP sensitized A549 and H1299 cells to erastin-induced ferroptosis through downregulation of NRF2; in addition, ectopic MT1DP upregulated malondialdehyde (MDA) and reactive oxygen species (ROS) levels, increased intracellular ferrous iron concentration, and reduced glutathione (GSH) levels in cancer cells exposed to erastin, whereas downregulation of MT1DP showed the opposite effect. RNA pulldown assay and dual-luciferase reporter assay confirmed that MT1DP modulated the expression of NRF2 via stabilizing miR-365a-3p. As low solubility of erastin limits its efficient application, we further prepared folate (FA)-modified liposome (FA-LP) nanoparticles for targeted co-delivery of erastin and MT1DP to enhance the bioavailability and the efficiency of the drug/gene combination. Erastin/MT1DP@FA-LPs (E/M@FA-LPs) sensitized erastin-induced ferroptosis with decreased cellular GSH levels and elevated lipid ROS. In vivo analysis showed that E/M@FA-LPs had a favorable therapeutic effect on lung cancer xenografts. In short, our findings identify a novel strategy to elevate erastin-induced ferroptosis in NSCLCs acting through the MT1DP/miR-365a-3p/NRF2 axis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Ferroptosis/drug effects , Liposomes/metabolism , Lung Neoplasms/drug therapy , NF-E2-Related Factor 2/drug effects , Piperazines/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Piperazines/pharmacology , Transfection
12.
Mol Cell Biochem ; 473(1-2): 217-228, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32642794

ABSTRACT

Ferroptosis is a newly discovered form of regulated cell death and characterized by an iron-dependent accumulation of lethal lipid reactive oxygen species (ROS), ferroptosis may exhibit a novel spectrum of clinical activity for cancer therapy. However, the significance of ferroptosis in the context of carcinoma biology is still emerging. Glycogen synthase kinase-3ß (GSK-3ß) has been found to be a fundamental element in weaking antioxidant cell defense by adjusting the nuclear factor erythroid 2-related factor 2 (Nrf2). In our study, decreased expression of GSK-3ß was observed in the cancer tissues of breast cancer patients, results of immunohistochemistry indicated that Nrf2 was highly expressed in low-GSK-3ß-expressed breast cancer tissues. The contributions of aberrant expression of GSK-3ß and Nrf2 to the erastin-induced ferroptosis in breast cancer were further assessed, silence of GSK-3ß blocked erastin-induced ferroptosis with less production of ROS and malondialdehyde (MDA) via upregulation of GPX4 and downregulation of arachidonate 15-lipoxygenase (Alox15), overexpression of GSK-3ß enhanced erastin-triggered ferroptosis with elevated ROS and MDA. Enhanced erastin-induced ferroptosis by overexpression of GSK-3ß was blocked by activating Nrf2. We further confirmed that overexpression of GSK-3ß strengthened erastin-induced tumor growth inhibition in breast cancer xenograft models in vivo. In summary, our findings conclude that modulation the balance between GSK-3ß/Nrf2 is a promising therapeutic approach and probably will be important targets to enhance the effect of erastin-induced ferroptosis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Ferroptosis/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , Piperazines/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Ferroptosis/genetics , Glycogen Synthase Kinase 3 beta/genetics , Humans , MCF-7 Cells , NF-E2-Related Factor 2/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics
13.
ACS Appl Bio Mater ; 3(5): 3345-3353, 2020 May 18.
Article in English | MEDLINE | ID: mdl-35025377

ABSTRACT

It is of great significance to design a multifunctional nanotherapeutic agent with flexible and intelligent response performance for a tumor microenvironment. Herein, hollow-structured hCu2-xS@Au derivatives were synthesized for multiswitchable drug release and synergistic photothermal therapy/chemotherapy. The remarkably improved release ratio (over 90%) under higher glutathione concentration and lower pH conditions could be ascribed to the fracture of the disulfide bond and the reduction of force between the drug and nanovehicles. In addition, the shrinking of tetradecanol molecules anchored on the surface of hCu2-xS@Au by near-infrared (NIR) photo-activation could also enhance the release of drugs in the cavity of hCu2-xS@Au. After decorating with Au nanoparticles, the favorable electron transitions endowed hCu2-xS@Au nanoparticles with a high photothermal conversion efficiency, which reaches 36.4% under a NIR laser irradiation of 808 nm. Compared to conventional single/double stimuli-responsive delivery, the proposed multimodal release pattern powerfully overcomes the limitation of insufficient dose as a single/double release unit for a tumor microenvironment was unable to meet the release requirements. After phagocytosis by tumor cells, the continuous release of anticancer drugs facilitated a substantial improvement in the antitumor therapy efficacy by combining chemo- and photothermal therapy in vivo. This design platform can provide a promising strategy for continuous and synergistic tumor therapy.

14.
J Cell Physiol ; 235(4): 3329-3339, 2020 04.
Article in English | MEDLINE | ID: mdl-31541463

ABSTRACT

Growing evidence confirms that ferroptosis plays an important role in tumor growth inhibition. However, some non-small-cell lung cancer (NSCLC) cell lines are less sensitive to erastin-induced ferroptotic cell death. Elucidating the mechanism of resistance of cancer cells to erastin-induced ferroptosis and increasing the sensitivity of cancer cells to erastin need to be addressed. In our experiment, erastin and acetaminophen (APAP) cotreatment inhibited NSCLC cell viability and promoted ferroptosis and apoptosis, accompanied with attenuation of glutathione and ectopic increases in lipid peroxides. Erastin and APAP promoted NSCLC cell death by regulating nucleus translocation of nuclear factor erythroid 2-related factor 2 (Nrf2); and the ferroptosis induced by erastin and APAP was abrogated by bardoxolone methyl (BM) with less generation of reactive oxygen species and malondialdehyde. As a downstream gene of Nrf2, heme oxygenase-1 expression decreased significantly with the cotreatment of erastin and APAP, which could be rescued by BM. In vivo experiment showed that the combination of erastin and APAP had a synergic therapeutic effect on xenograft of lung cancer. In short, the present study develops a new effective treatment for NSCLC by synergizing erastin and APAP to induce ferroptosis.


Subject(s)
Acetaminophen/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Heme Oxygenase-1/genetics , NF-E2-Related Factor 2/genetics , Piperazines/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Ferroptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lipid Peroxides/metabolism , Malondialdehyde/metabolism , Mice , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Cancer Sci ; 110(10): 3173-3182, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31464035

ABSTRACT

Ferroptosis is an iron-dependent, lipid peroxide-driven cell death caused by inhibition of the cystine/glutamate transporter, which is of importance for the survival of triple-negative breast cancer (TNBC) cells. Erastin is a low molecular weight chemotherapy drug that induces ferroptosis; however, poor water solubility and renal toxicity have limited its application. Exosomes, as drug delivery vehicles with low immunogenicity, high biocompatibility and high efficiency, have attracted increasing attention in recent years. Herein, we developed a formulation of erastin-loaded exosomes labeled with folate (FA) to form FA-vectorized exosomes loaded with erastin (erastin@FA-exo) to target TNBC cells with overexpression of FA receptors. The characterization, drug release, internalization and anti-tumor effect in vitro of erastin@FA-exo were determined. Erastin@FA-exo could increase the uptake efficiency of erastin into MDA-MB-231 cells; compared with erastin@exo and free erastin, erastin@FA-exo has a better inhibitory effect on the proliferation and migration of MDA-MB-231 cells. Furthermore, erastin@FA-exo promoted ferroptosis with intracellular depletion of glutathione and reactive oxygen species overgeneration. Western blot analyses revealed that erastin@FA-exo suppressed expression of glutathione peroxidase 4 (GPX4) and upregulated expression of cysteine dioxygenase (CDO1). We conclude that targeting and biocompatibility of exosome-based erastin preparations provide an innovative and powerful delivery platform for anti-cancer therapy.


Subject(s)
Exosomes/chemistry , Folic Acid/chemistry , Piperazines/pharmacology , Triple Negative Breast Neoplasms/metabolism , Cell Death , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cysteine Dioxygenase/metabolism , Drug Delivery Systems , Gene Expression Regulation, Neoplastic/drug effects , Glutathione Peroxidase/metabolism , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase , Piperazines/chemistry , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/drug therapy
16.
Front Pharmacol ; 10: 75, 2019.
Article in English | MEDLINE | ID: mdl-30787876

ABSTRACT

Combination therapy which enhances efficacy and reduces toxicity, has been increasingly applied as a promising strategy for cancer therapy. Here, a reactive oxygen species (ROS) that enhanced combination chemotherapy nanodevices was fabricated based on the Fe-chelated polydopamine (PDA) nanoparticles (NPs). The structure was characterized by dynamic light scattering-autosizer, transmission electron microscopy, energy dispersive spectroscopy, and Fourier-transform infrared (FT-IR) spectrophotometer. The in vitro drug release profile triggered by low intracellular pH indicated that the system demonstrated controlled therapeutic activity. In vitro cell uptake studies showed that doxorubicin (DOX)-loaded Fe-PDA/ folic acid (FA)- polyethylene glycol (DOX@Fe-PDA/FA-PEG) had a strong uptake capacity and can be rapidly internalized by MCF-7 cells. The in vitro experiments demonstrated that DOX@Fe-PDA/FA-PEG triggered the intracellular ROS overproduction, thereby enhancing its therapeutic effect on breast cancer. In summary, this experiment demonstrated the novel DOX-loaded composite NPs used as a potential targeted nanocarrier for breast cancer treatment, which could be a promising therapeutic strategy against breast cancer.

17.
Front Oncol ; 8: 507, 2018.
Article in English | MEDLINE | ID: mdl-30450337

ABSTRACT

Ferroptosis is a type of programmed cell death characterized by the accumulation of lipid reactive oxygen species (L-ROS) driven by the oxidative degeneration of lipids in an iron-dependent manner. The mechanism by which lipid oxidative degradation drives ROS-ferroptosis involves metabolic dysfunctions that result in impaired intracellular metabolic processes and ROS production. Recent studies have found that p53 acts as a positive regulator of ferroptosis by promoting ROS production. p53 directly regulates the metabolic versatility of cells by favoring mitochondrial respiration, leading to ROS-mediated ferroptosis. In mild stress, p53 protects cell survival via eliminating ROS; additionally, in human colorectal cancer, p53 antagonizes ferroptosis by formation of the DPP4-p53 complex. In short, the mechanisms of p53-mediated ROS production underlying cellular response are poorly understood. In the context of recent research results, the indistinct roles of p53 on ROS-mediated ferroptosis are scrutinized to understand the mechanism underlying p53-mediated tumor suppression.

SELECTION OF CITATIONS
SEARCH DETAIL
...