Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Nano ; 18(33): 22257-22274, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39121010

RESUMEN

Iron metabolism has emerged as a promising target for cancer therapy; however, the innate metabolic compensatory capacity of cancer cells significantly limits the effectiveness of iron metabolism therapy. Herein, bioactive gallium sulfide nanodots (GaSx), with dual functions of "reprogramming" and "interfering" iron metabolic pathways, were successfully developed for tumor iron metabolism therapy. The constructed GaSx nanodots ingeniously harness hydrogen sulfide (H2S) gas, which is released in response to the tumor microenvironment, to reprogram the inherent transferrin receptor 1 (TfR1)-ferroportin 1 (FPN1) iron metabolism axis in cancer cells. Concurrently, the gallium ions (Ga3+) derived from GaSx act as a biochemical "Trojan horse", mimicking the role of iron and displacing it from essential biomolecular binding sites, thereby influencing the fate of cancer cells. By leveraging the dual mechanisms of Ga3+-mediated iron disruption and H2S-facilitated reprogramming of iron metabolic pathways, GaSx prompted the initiation of a paraptosis-apoptosis hybrid pathway in cancer cells, leading to marked suppression of tumor proliferation. Importantly, the dysregulation of iron metabolism induced by GaSx notably increased tumor cell susceptibility to both chemotherapy and immune checkpoint blockade (ICB) therapy. This study underscores the therapeutic promise of gas-based interventions and metal ion interference strategies for the tumor metabolism treatment.


Asunto(s)
Apoptosis , Galio , Hierro , Humanos , Galio/química , Galio/farmacología , Apoptosis/efectos de los fármacos , Hierro/metabolismo , Hierro/química , Animales , Ratones , Proliferación Celular/efectos de los fármacos , Receptores de Transferrina/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Sulfuros/química , Sulfuros/farmacología , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Paraptosis , Proteínas de Transporte de Catión
2.
Front Psychol ; 15: 1412034, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38988398

RESUMEN

This study integrates SOR (Stimuli-Organism-Response) theoretical framework and rational behavior theory within a theoretical framework, incorporating group norms as a moderating factor to investigate the psychological mechanisms influencing Chinese college students' online rumor-refutation behavior amidst public health crises. Using the structural equation modeling research method, data was collected via questionnaires from 1,254 participants in the context of the COVID-19 pandemic. The findings indicate that both online and offline information seeking are positively correlated with college students' attitudes and subjective norms. Moreover, the attitudes and subjective norms of college students are positively correlated with the online rumor refuting behavior. Furthermore, group norms serve to strengthen the connection between college students' attitudes and their engagement in online refuting rumors. These results illuminate the psychological underpinnings driving college students' online rumor-refuting actions, offering practical and policy implications for effectively managing rumor behaviors.

3.
Biochem Biophys Res Commun ; 730: 150366, 2024 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-38991254

RESUMEN

Laryngeal squamous cell carcinoma (LSCC) with a high incidence and mortality rate, has a serious impact worldwide. Phosphofructokinase-1 liver type (PFKL) is a major enzyme in glycolysis progress, but its role in modulating tumorigenesis and cisplatin (DDP) chemosensitivity of LSCC was still unclear. The mRNA and protein levels of PFKL were detected by qRT-PCR and immunohistochemical assay. Cell Counting Kit-8 assay and flow cytometry were carried out to observe cell viability, as well as apoptosis and mitochondrial reactive oxygen species (mito-ROS). Extracellular acidification rate and lactate content were measured using extracellular flux analysis and lactate assay kit. Immunofluorescent staining was used to evaluate the expression of γ-H2AX foci. DNA damage was detected via single-cell gel electrophoresis. Western blotting was introduced to evaluate the protein level of PFKL, LDHA, γ-H2AX, cleaved PARP, H3K27ac, and H3K9ac. Mice xenograft model of LSCC was built for in vivo validation. The PFKL expression was significantly increased in LSCC and associated with poor survival of LSCC patients. Knockdown of PFKL in LSCC cells significantly inhibited cell viability, ECAR, lactate content, and LDHA expression, but promoted mito-ROS level. Furthermore, knockdown of PFKL enhanced response of LSCC cells to DDP by increasing DDP-induced apoptosis, promoting DDP-induced mito-ROS level, γ-H2AX foci, tail DNA, and the expression of γ-H2AX and cleaved PARP. However, the overexpression of PFKL in LSCC cells had opposite experimental results. Nude mice tumor formation experiment proved that downregulation of PFKL significantly enhanced response of cells to DDP, demonstrated by reduced tumor volume, weight and increased TUNEL-positive cells. Suppression of CBP/EP300-mediated PFKL transcription inhibited cell viability and glycolysis and promoted mito-ROS in LSCC. PFKL promotes cell viability and DNA damage repair in DDP-treated LSCC through regulation of glycolysis pathway.


Asunto(s)
Antineoplásicos , Supervivencia Celular , Cisplatino , Glucólisis , Neoplasias Laríngeas , Ratones Desnudos , Cisplatino/farmacología , Glucólisis/efectos de los fármacos , Humanos , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/tratamiento farmacológico , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/genética , Animales , Supervivencia Celular/efectos de los fármacos , Ratones , Línea Celular Tumoral , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Fosfofructoquinasa-1/metabolismo , Fosfofructoquinasa-1/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Ratones Endogámicos BALB C , Daño del ADN/efectos de los fármacos
4.
ACS Nano ; 18(20): 12830-12844, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38709246

RESUMEN

The immunosuppressive microenvironment of cervical cancer significantly hampers the effectiveness of immunotherapy. Herein, PEGylated manganese-doped calcium sulfide nanoparticles (MCSP) were developed to effectively enhance the antitumor immune response of the cervical cancer through gas-amplified metalloimmunotherapy with dual activation of pyroptosis and STING pathway. The bioactive MCSP exhibited the ability to rapidly release Ca2+, Mn2+, and H2S in response to the tumor microenvironment. H2S disrupted the calcium buffer system of cancer cells by interfering with the oxidative phosphorylation pathway, leading to calcium overload-triggered pyroptosis. On the other hand, H2S-mediated mitochondrial dysfunction further promoted the release of mitochondrial DNA (mtDNA), enhancing the activation effect of Mn2+ on the cGAS-STING signaling axis and thereby activating immunosuppressed dendritic cells. The released H2S acted as an important synergist between Mn2+ and Ca2+ by modulating dual signaling mechanisms to bridge innate and adaptive immune responses. The combination of MCSP NPs and PD-1 immunotherapy achieved synergistic antitumor effects and effectively inhibited tumor growth. This study reveals the potential collaboration between H2S gas therapy and metalloimmunotherapy and provides an idea for the design of nanoimmunomodulators for rational regulation of the immunosuppressive tumor microenvironment.


Asunto(s)
Inmunoterapia , Proteínas de la Membrana , Piroptosis , Microambiente Tumoral , Neoplasias del Cuello Uterino , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/terapia , Femenino , Humanos , Ratones , Animales , Piroptosis/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Manganeso/química , Manganeso/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Nanopartículas/química , Transducción de Señal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Calcio/metabolismo , Ratones Endogámicos BALB C , Ensayos de Selección de Medicamentos Antitumorales
5.
ACS Nano ; 18(15): 10542-10556, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38561324

RESUMEN

Immunotherapy has emerged as a potential approach for breast cancer treatment. However, the rigid stromal microenvironment and low immunogenicity of breast tumors strongly reduce sensitivity to immunotherapy. To sensitize patients to breast cancer immunotherapy, hyaluronic acid-modified zinc peroxide-iron nanocomposites (Fe-ZnO2@HA, abbreviated FZOH) were synthesized to remodel the stromal microenvironment and increase tumor immunogenicity. The constructed FZOH spontaneously generated highly oxidative hydroxyl radicals (·OH) that degrade hyaluronic acid (HA) in the tumor extracellular matrix (ECM), thereby reshaping the tumor stromal microenvironment and enhancing blood perfusion, drug penetration, and immune cell infiltration. Furthermore, FZOH not only triggers pyroptosis through the activation of the caspase-1/GSDMD-dependent pathway but also induces ferroptosis through various mechanisms, including increasing the levels of Fe2+ in the intracellular iron pool, downregulating the expression of FPN1 to inhibit iron efflux, and activating the p53 signaling pathway to cause the failure of the SLC7A11-GSH-GPX4 signaling axis. Upon treatment with FZOH, 4T1 cancer cells undergo both ferroptosis and pyroptosis, exhibiting a strong immunogenic response. The remodeling of the tumor stromal microenvironment and the immunogenic response of the cells induced by FZOH collectively compensate for the limitations of cancer immunotherapy and significantly enhance the antitumor immune response to the immune checkpoint inhibitor αPD-1. This study proposes a perspective for enhancing immune therapy for breast cancer.


Asunto(s)
Neoplasias de la Mama , Neoplasias , Humanos , Femenino , Neoplasias de la Mama/terapia , Ácido Hialurónico , Inmunoterapia , Peróxidos , Zinc , Microambiente Tumoral , Línea Celular Tumoral
6.
J Colloid Interface Sci ; 662: 857-869, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38382370

RESUMEN

Copper-based nanomaterials show considerable potential in the chemodynamic therapy of cancers. However, their clinical application is restricted by low catalytic activity in tumor microenvironment and copper-induced tumor angiogenesis. Herein, a novel copper-doxorubicin-anlotinib (CDA) nanoconjugate was constructed by the combination of copper-hydrazide coordination, hydrazone linkage and Schiff base bond. The CDA nanoconjugate consists of a copper-3,3'-dithiobis(propionohydrazide)-doxorubicin core and an anlotinib-hyaluronan shell. Benefiting from hyaluronan camouflage and abundant disulfide bonds and Cu2+, the CDA nanoconjugate possessed excellent tumor-targeting and glutathione-depleting abilities and enhanced chemodynamic efficacy. Released doxorubicin significantly improved copper-mediated chemodynamic therapy by upregulating nicotinamide adenine dinucleotide phosphate oxidase 4 expression to increase intracellular H2O2 level. Furthermore, the nanoconjugate produced excessive •OH to induce lipid peroxidation and mitochondrial dysfunction, thus greatly elevating doxorubicin-mediated chemotherapy. Importantly, anlotinib effectively inhibited the angiogenic potential of copper ions. In a word, the CDA nanoconjugate is successfully constructed by combined coordination and pH-responsive linkages, and displays the great potential of copper-drug conjugate for targeted synergistic chemo/chemodynamic/antiangiogenic triple therapy against cancers.


Asunto(s)
Carcinoma Hepatocelular , Indoles , Neoplasias Hepáticas , Nanopartículas , Neoplasias , Quinolinas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Cobre , Ácido Hialurónico , Nanoconjugados , Peróxido de Hidrógeno , Neoplasias Hepáticas/tratamiento farmacológico , Doxorrubicina/farmacología , Glutatión , Línea Celular Tumoral , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA