Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Nano ; 17(20): 20218-20236, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37838975

ABSTRACT

Low-temperature photothermal therapy (PTT) is a noninvasive method that harnesses the photothermal effect at low temperatures to selectively eliminate tumor cells, while safeguarding normal tissues, minimizing thermal damage, and enhancing treatment safety. First we evaluated the transcriptome of tumor cells at the gene level following low-temperature treatment and observed significant enrichment of genes involved in cell cycle and heat response-related signaling pathways. To address this challenge, we have developed an engineering multifunctional nanoplatform that offered an all-in-one strategy for efficient sensitization of low-temperature PTT. Specifically, we utilized MoS2 nanoparticles as the photothermal core to generate low temperature (40-48 °C). The nanoplatform was coated with DPA to load CPT-11 and Fe2+ and was further modified with PEG and iRGD to enhance tumor specificity (MoS2/Fe@CPT-11-PEG-iRGD). Laser- and acid-triggered release of CPT-11 can significantly increase intracellular H2O2 content, cooperate with Fe2+ ions to increase intracellular lipid ROS content, and activate ferroptosis. Furthermore, CPT-11 induced cell cycle arrest in the temperature-sensitive S-phase, and increased lipid ROS levels contributed to the degradation of HSPs protein expression. This synergistic approach could effectively induce tumor cell death by the sensitized low-temperature PTT and the combination of ferroptosis and chemotherapy. Our nanoplatform can also maximize tumor cell eradication and prolong the survival time of tumor-bearing mice in vivo. The multifunctional approach will provide more possibilities for clinical applications of low-temperature PTT and potential avenues for the development of multiple tumor treatments.


Subject(s)
Nanoparticles , Neoplasms , Animals , Mice , Temperature , Photothermal Therapy , Irinotecan/therapeutic use , Molybdenum/therapeutic use , Reactive Oxygen Species/therapeutic use , Hydrogen Peroxide , Neoplasms/therapy , Lipids , Phototherapy/methods , Cell Line, Tumor
2.
ACS Nano ; 17(5): 4667-4687, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36861638

ABSTRACT

The hypoxia microenvironment of solid tumors poses a technological bottleneck for ferroptosis and immunotherapy in clinical oncology. Nanoreactors based on special physiological signals in tumor cells are able to avoid various tumor tolerance mechanisms by alleviating the intracellular hypoxia environment. Herein we reported a nanoreactor Cu2-xSe that enabled the conversion of Cu elements between Cu+ and Cu2+ for the generation of O2 and the consumption of intracellular GSH content. Furthermore, to enhance the catalytic and ferroptosis-inducing activities of the nanoreactors, the ferroptosis agonist Erastin was loaded on the ZIF-8 coating on the surface of Cu2-xSe to up-regulate the expression of NOX4 protein, increase the intracellular H2O2 content, catalyze the Cu+ to produce O2 and activate ferroptosis. In addition, the nanoreactors were simultaneously surface functionalized with PEG polymer and folic acid molecules, which ensured the in vivo blood circulation and tumor-specific uptake. In vitro and in vivo experiments demonstrated that the functionalized self-supplying nanoreactors can amplify the ability to generate O2 and consume intracellular GSH via the interconversion of Cu elements Cu+ and Cu2+, and impair the GPX4/GSH pathway and HIF-1α protein expression. At the same time, by alleviating the intracellular hypoxia environment, the expression of miR301, a gene in the secreted exosomes was decreased, which ultimately affected the phenotype polarization of TAMs and increased the content of IFN γ secreted by CD8+ T cells, which further promoted the ferroptosis induced by Erastin-loaded nanoreactors. This combined therapeutic strategy of activating the tumor immune response and ferroptosis via self-supplying nanoreactors provides a potential strategy for clinical application.


Subject(s)
Ferroptosis , Neoplasms , Triple Negative Breast Neoplasms , Humans , Oxygen , Triple Negative Breast Neoplasms/drug therapy , CD8-Positive T-Lymphocytes , Hydrogen Peroxide , Immunotherapy , Hypoxia , Copper , Nanotechnology , Cell Line, Tumor , Tumor Microenvironment
3.
ACS Nano ; 16(2): 2381-2398, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35041395

ABSTRACT

Ferroptosis is a recently discovered route of regulated cell death that offers the opportunities for the treatment of chemotherapy-resistant tumor indications, but its efficacy can be affected by the glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1) antioxidant mechanisms, posing significant challenges for its clinical translation. In this study, we report a Cu-tetra(4-carboxyphenyl)porphyrin chloride(Fe(III)) (Cu-TCPP(Fe)) metal organic framework (MOF)-based nanosystem for the efficient incorporation of Au nanoparticles (NPs) and RSL3, which can demonstrate enzyme-like activities to universally suppress the antiferroptotic pathways in tumor cells for amplifying ferroptotic damage. Herein, Cu-TCPP(Fe) MOF nanosheets were integrated with Au NPs via in situ nucleation and loaded with RSL3 via π-π stacking, which were eventually modified with polyethylene glycol (PEG) and iRGD for tumor-targeted drug delivery. Specifically, the Au NPs can demonstrate glucose oxidase-like activities for efficient glucose depletion, thus disrupting the pentose phosphate pathway to impede reduced glutathione (GSH) biosynthesis and prevent the recycling of coenzyme Q10 (CoQ10) to CoQ10H2, while Cu species can oxidize GSH into oxidized glutathione (GSSG). These nanocatalytic activities can lead to the simultaneous inhibition of the GPX4/GSH and FSP1/CoQ10H2 pathways and cooperate with the GPX4-deactivating function of RSL3 to cause pronounced ferroptotic damage, thereby providing a strong rationale for the application of ferroptosis therapy in the clinic.


Subject(s)
Ferroptosis , Metal Nanoparticles , Triple Negative Breast Neoplasms , Ferric Compounds , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Gold/pharmacology , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase , Triple Negative Breast Neoplasms/drug therapy
4.
Small ; 17(40): e2102046, 2021 10.
Article in English | MEDLINE | ID: mdl-34448349

ABSTRACT

Liver tumor is difficult to cure for its high degree of malignancy and rapid progression characteristics. Ferroptosis as a new model of inducing cell death is expected to break the treatment bottleneck of liver tumors. Here, a strategy to induce ferroptosis in HepG2 cells with acid-degradable tumor targeted nanosheets Cu-Hemin-PEG-Lactose acid (Cu-Hemin-PEG-LA) is proposed. After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. Furthermore, the expression of glutathione peroxidase 4 protein (GPX4) is down-regulated by consumption intracellular GSH content via converting GSH into glutathione oxidized (GSSG), which is named the classical mode. The intracellular Fe2+ content is overloaded by the significant up-regulation of HMOX1 expression, which is denoted as nonclassical mode. The synergistic effect of classical and nonclassical mode increased the intracellular lipid reactive oxide species, induced the occurrence of ferroptosis and up-regulated the expression of BH3 interacting domain death agonist (BID), apoptosis-inducing factor (AIF), and endonuclease G proteins (EndoG). The synergistic strategy demonstrate the excellent ferroptosis induction ability and antitumor efficacy in vivo, which provides great potential for the clinical transformation of ferroptosis.


Subject(s)
Ferroptosis , Liver Neoplasms , Ferrous Compounds , Glutathione , Hep G2 Cells , Humans
5.
ACS Nano ; 14(10): 14164-14180, 2020 10 27.
Article in English | MEDLINE | ID: mdl-32975406

ABSTRACT

As an increased product of high-rate aerobic glycolysis in tumors, lactate could regulate the immunosuppressive tumor microenvironment (TME). A PEG-CDM surface modified, GSH-dependent responsive hollow mesoporous organosilica nanoplatform loaded with hydroxycamptothecin (HCPT) and siMCT-4 was administrated for synergistic tumor chemo-immunotherapy. The nanoplatform cascaded responded to the weak acid TME and the high level of GSH in tumor cells. HCPT and siMCT-4 were continuously released from the nanoplatform for chemotherapy and inhibiting intracellular lactate efflux. The increased intracellular lactate and HCPT effectively induced tumor cell apoptosis. Moreover, the decreased extracellular lactate polarized tumor-associated macrophages (TAMs) phenotype from M2 type to M1 type and restored CD8+ T cell activity in vivo. The results demonstrated that the nanoplatform effectively removed the immunosuppressive TME, inhibited tumor growth, and suppressed lung metastasis of B16F10 cells and 4T1 cells via the combination of inhibiting lactate efflux and chemotherapy. Accordingly, it suggested a strategy to transform immunosuppressive tumors into "hot" tumors and inhibit the tumor growth with high efficiency in vivo.


Subject(s)
Nanoparticles , Drug Delivery Systems , Immunotherapy , Lactic Acid , Tumor Microenvironment
6.
Therap Adv Gastroenterol ; 13: 1756284820911259, 2020.
Article in English | MEDLINE | ID: mdl-32231711

ABSTRACT

BACKGROUND: To date, the prognostic significance of acellular mucin pools in tumors from patients with locally advanced rectal cancer (LARC) undergoing preoperative chemoradiotherapy (CRT) and subsequently obtaining pathological complete response (pCR) has not been well determined. Our current study aimed to explore the prognostic impact on these patients of acellular mucin pools. METHODS: We collected clinical data from 117 consecutive LARC patients who achieved pCR after preoperative CRT and then underwent radical resection. Two groups of patients were generated, according to the presence or absence of acellular mucin pools. The 5-year disease-free survival (DFS) and overall survival (OS) rates were compared between the two groups of patients. RESULTS: A total of 27 (23.1%) patients presented with acellular mucin pools. At a median follow-up period of 64 months, patients with acellular mucin pool showed a 5-year DFS rate (96.3% versus 83.7%, p = 0.110) and 5-year OS rate (100% versus 87.5%, p = 0.054) statistically similar to those of patients without acellular mucin pools. In univariable and multivariable Cox regression analyses, the presence of acellular mucin pools was not determined as an independent risk factor for DFS [hazard ratio (HR): 0.222; 95% confidence interval (CI): 0.029-1.864; p = 0.145] or OS (HR: 0.033; 95% CI: 0.000-9.620; p = 0.238). CONCLUSIONS: Acellular mucin pools had no significant prognostic impact on LARC patients showing pCR after preoperative CRT.

7.
Biochem Biophys Res Commun ; 526(2): 321-327, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32220496

ABSTRACT

The chemotherapeutic efficacy of paclitaxel against hypoxic tumors is usually unsatisfactory, which is partially due to the so-called hypoxia-induced drug resistance. The mechanism of hypoxia-induced resistance is primarily associated with hypoxia-inducible factor 1α (HIF-1α), which is an oxygen-sensitive transcriptional activator coordinating the cellular response to hypoxia. Apigenin is a natural occurring HIF-1α inhibitor that can suppress the expression of HIF-1α through multiple pathways and reverse the hypoxia-induced resistance found in cancer cells. Here we report that the use of apigenin can suppress the HIF-1α expression in hypoxic tumors through the simultaneous inhibition of the AKT/p-AKT pathway and HSP90, which is beneficial for enhancing the anticancer activity of the co-administered paclitaxel. The potential synergistic effect of apigenin and paclitaxel was further validated on HepG2 cell line and tumor-bearing mouse models.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apigenin/pharmacology , Cell Hypoxia/drug effects , Drug Resistance, Neoplasm/drug effects , Liver Neoplasms/drug therapy , Paclitaxel/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apigenin/administration & dosage , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Injections, Intravenous , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/administration & dosage , Structure-Activity Relationship , Tumor Cells, Cultured
8.
J Environ Manage ; 260: 110130, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-31941638

ABSTRACT

Methods of removing phosphate from wastewater with a low phosphate concentration are of great environmental significance. In this study, immobilized beads were prepared by entrapping modified bentonite powder in calcium-alginate (Al-NaBT-CA), and the potential of the beads for phosphate removal from wastewater was investigated. The effects of pH (1-10) and initial phosphate concentration (0.5-50 mg/L) were also examined in batch experiments with Al-NaBT-CA beads. The optimum pH value for phosphate removal by Al-NaBT-CA beads was pH 3. In addition, a high initial phosphate concentration promoted phosphate adsorption. Adsorption kinetics showed that the adsorption of phosphate using beads followed a pseudo-second-order kinetic model (R2 = 0.98-0.99). The adsorption isotherm data was well fitted by the Sips adsorption model. The maximum phosphate adsorption capacity of the Al-NaBT-CA beads was 15.77 mg/g, which was slightly less than that of the modified powder. The specific surface area of the Al-NaBT-CA beads was 17.01 m2/g, and their average pore size was 13.41 nm. Scanning electron microscopy suggested that the high inner porosity and rough outer surface of the beads facilitated phosphate transfer.


Subject(s)
Wastewater , Water Pollutants, Chemical , Adsorption , Alginates , Bentonite , Glucuronic Acid , Hexuronic Acids , Hydrogen-Ion Concentration , Kinetics , Phosphates
9.
Nanoscale ; 12(1): 130-144, 2020 Jan 07.
Article in English | MEDLINE | ID: mdl-31799577

ABSTRACT

Tumor-associated macrophages (TAMs) are the most important components in the tumor immunosuppressive microenvironment, promoting tumor growth and metastasis. Although TAMs have become one of the hot topics of tumor immunotherapy, challenges still remain to achieve TAM-targeted re-polarization therapy. In this work, porous hollow iron oxide nanoparticles (PHNPs) were synthesized for loading a P13K γ small molecule inhibitor (3-methyladenine, 3-MA) and further modified by mannose to target TAMs. The delivery system named PHNPs@DPA-S-S-BSA-MA@3-MA showed good efficiency for targeting TAMs. The inflammatory factor NF-κB p65 of macrophages was activated by the combination of PHNPs and 3-MA, which synergistically switched TAMs to pro-inflammatory M1-type macrophages. As a result, it activated immune responses and inhibited tumor growth in vivo. The study provides an intracellular switch of the TAM phenotype for targeted TAM therapy.


Subject(s)
Ferrosoferric Oxide/chemistry , Macrophages/immunology , Nanoparticles/chemistry , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line , Cell Survival/drug effects , Drug Carriers/chemistry , Female , Humans , Immunotherapy , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Polyethylene Glycols/chemistry , Porosity , Serum Albumin, Bovine/chemistry , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...