Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Gut Microbes ; 16(1): 2307542, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38319728

RESUMEN

The gut microbiota and Short-chain fatty acids (SCFAs) can influence the progression of diseases, yet the role of these factors on gastric cancer (GC) remains uncertain. In this work, the analysis of the gut microbiota composition and SCFA content in the blood and feces of both healthy individuals and GC patients indicated that significant reductions in the abundance of intestinal bacteria involved in SCFA production were observed in GC patients compared with the controls. ABX mice transplanted with fecal microbiota from GC patients developed more tumors during the induction of GC and had lower levels of butyric acid. Supplementation of butyrate during the induction of gastric cancer along with H. pylori and N-methyl-N-nitrosourea (MNU) in WT in GPR109A-/-mice resulted in fewer tumors and more IFN-γ+ CD8+ T cells, but this effect was significantly weakened after knockout of GPR109A. Furthermore, In vitro GC cells and co-cultured CD8+ T cells or CAR-Claudin 18.2+ CD8+ T cells, as well as in vivo tumor-bearing studies, have indicated that butyrate enhanced the killing function of CD8+ T cells or CAR-Claudin 18.2+ CD8+ T cells against GC cells through G protein-coupled receptor 109A (GPR109A) and homologous domain protein homologous box (HOPX). Together, these data highlighted that the restoration of gut microbial butyrate enhanced CD8+ T cell cytotoxicity via GPR109A/HOPX, thus inhibiting GC carcinogenesis, which suggests a novel theoretical foundation for GC management against GC.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias Gástricas , Humanos , Ratones , Animales , Butiratos/metabolismo , Microbioma Gastrointestinal/fisiología , Linfocitos T CD8-positivos , Ácidos Grasos Volátiles/metabolismo , Ácido Butírico , Claudinas
2.
Int J Nanomedicine ; 17: 4659-4675, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36199474

RESUMEN

Purpose: Lymphadenectomy with lymph node (LN) mapping is essential for surgical removal of solid tumors. Existing agents do not provide accurate multimodal mapping and antitumor therapy for metastatic LNs; therefore, we fabricated a polydopamine (PDA) nanoparticle (NP)-based tumor-targeted LN mapping agent capable of multimodal mapping and guided photothermal therapy (PTT) for metastatic LNs. Materials and Methods: PDA NPs modified with polyethylene glycol (PEG) were obtained by polymerization under alkaline conditions. The PEG-PDA NPs were loaded with the circular tripeptide Arg-Gly-Asp (cRGD) to achieve tumor-targeting capacity and with the fluorescent dye IR820 and magnetic resonance imaging (MRI) contrast Gd(NH2)2 for in situ detection. The resulting cRGD-PEG-PDA@IR820/Gd(NH2)2 (cRGD-PPIG) NPs were tested for their biosafety and metastatic LN mapping ability. They were drained specifically into LNs and selectively taken up by gastric MKN45 cells via αvß3 integrin-mediated endocytosis. Results: This phenomenon enabled MR/optical/near-infrared fluorescence multimodal metastatic LN mapping, guiding the creation of accurate and highly efficient PTT for gastric cancer metastatic LNs in mice. Conclusion: In summary, we fabricated tumor-targeted cRGD-PPIG NPs with MR/optical/near-infrared fluorescence multimodal metastatic LN mapping capacity for surgery and efficient PTT guidance post-surgery.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias Gástricas , Animales , Línea Celular Tumoral , Medios de Contraste , Colorantes Fluorescentes , Indoles , Integrina beta3 , Ganglios Linfáticos/diagnóstico por imagen , Ratones , Fototerapia/métodos , Terapia Fototérmica , Polietilenglicoles , Polímeros
3.
Biomater Adv ; 138: 212919, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35913232

RESUMEN

Photothermal therapy (PTT) usually causes hyperthermia and damages healthy tissues. Developing a PTT platform with enhanced therapeutic effects and reduced side effects to normal tissues attracts increasing attention. Herein, we developed a multifunctional theranostic nanoplatform using poly(lactic-co-glycolic acid) (PLGA) loaded with near-infrared (NIR) photothermal agent (new indocyanine green IR820), fluorescence imaging agent (ZnCdSe/ZnS quantum dots, QDs) and autophagy inhibitor (chloroquine, CQ). These PLGA/IR820/Fluorescence imaging agent/CQ co-loading nanoparticles (termed PIFC NPs) displayed photothermal effects, enhanced the stability of IR820 in vivo, and enabled QDs to have stable fluorescent signals in vitro and in vivo. The PIFC NPs with particle size around 240 nm aggregated to tumor sites through the high permeability and retention effects of solid tumors. The intracellular delivery of CQ molecules through PIFC NPs significantly attenuated the degradation of autophagic lysosomes in tumor cells and effectively inhibited the autophagy mediated repair of photothermal damaged cells. Under milder NIR irradiation conditions, PIFC NPs exhibited high antitumor effect. By regulating autophagy, PTT can be effectively sensitized, which will provide a new idea for future cancer treatment research.


Asunto(s)
Hipertermia Inducida , Neoplasias , Autofagia , Humanos , Neoplasias/terapia , Fototerapia/métodos , Terapia Fototérmica , Medicina de Precisión
4.
Drug Deliv ; 29(1): 161-173, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34967262

RESUMEN

A major impediment in the development of nanoplatform-based ovarian cancer therapy is endo/lysosome entrapment. To solve this dilemma, a hollow mesoporous organosilica-based nanoplatform (HMON@CuS/Gd2O3) with a mild-temperature photothermal therapeutic effect and multimodal imaging abilities was successfully synthesized. HMON@CuS/Gd2O3 exhibited an appropriate size distribution, L-glutathione (GSH)-responsive degradable properties, and high singlet oxygen generation characteristics. In this study, the nanoplatform specifically entered SKOV-3 cells and was entrapped in endo/lysosomes. With a mild near infrared (NIR) power density (.5 W/cm2), the HMON@CuS/Gd2O3 nanoplatform caused lysosome vacuolation, disrupted the lysosomal membrane integrity, and exerted antitumour effects in ovarian cancer. Additionally, our in vivo experiments indicated that HMON@CuS/Gd2O3 has enhanced T1 MR imaging, fluorescence (FL) imaging (wrapping fluorescent agent), and infrared thermal (IRT) imaging capacities. Using FL/MRI/IRT imaging, HMON@CuS/Gd2O3 selectively caused mild phototherapy in the cancer region, efficiently inhibiting the growth of ovarian cancer without systemic toxicity in vivo. Taken together, the results showed that these well-synthesized nanoplatforms are likely promising anticancer agents to treat ovarian cancer and show great potential for biomedical applications.


Asunto(s)
Endosomas/efectos de los fármacos , Compuestos de Organosilicio/química , Neoplasias Ováricas/patología , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Química Farmacéutica , Femenino , Humanos , Concentración de Iones de Hidrógeno , Imagen Multimodal , Propiedades de Superficie
5.
Clin Transl Immunology ; 10(9): e1338, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34584694

RESUMEN

OBJECTIVE: The benefit of Se supplementation in rheumatoid arthritis (RA) has been tested in clinical trials, but results remain inconclusive. The objective of this study was to specifically investigate the potential benefit of supranutritional Se by examining human samples from an area with supranutritional Se intake and testing a mouse model of RA. METHODS: Peripheral blood mononuclear cells (PBMCs) from RA patients (N = 57) and healthy controls (HC, N = 71) from an area of supranutritional Se intake (Enshi, Hubei, China) were analysed by flow cytometry. Serum cytokine and Se levels were measured by cytometric beads array (CBA) and inductively coupled plasma mass spectrometry (ICP-MS), respectively. With sufficient or supranutritional selenium intake, mice were induced with collagen-induced arthritis (CIA) and examined for disease activity and immunopathology. The influence of Se supplementation in the generation of RANKL-expressing osteoclastogenic CD4+ T cells was investigated by in vitro assays. RESULTS: In Enshi city, HC showed the above-normal concentrations of serum Se concentrations while RA patients were enriched in the normal range (70-150 ng mL-1) or below. RA patients with higher Se levels demonstrated milder disease and lower levels of C-reactive protein, IL-6, RANKL and Th17 cells. In the mouse CIA model, supranutritional Se supplementation delayed disease onset, ameliorated joint pathology and reduced CD4+CD44+RANKL+ T cells. Se supplementation could suppress RANKL expression in cultured mouse Th17 cells. CONCLUSION: Supranutritional Se suppresses RANKL-expressing osteoclastogenic CD4+ T cells and could be beneficial to RA, which warrants formal testing in randomised clinical trials.

6.
Nat Immunol ; 22(9): 1127-1139, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34413521

RESUMEN

Follicular helper T (TFH) cells are a specialized subset of CD4+ T cells that essentially support germinal center responses where high-affinity and long-lived humoral immunity is generated. The regulation of TFH cell survival remains unclear. Here we report that TFH cells show intensified lipid peroxidation and altered mitochondrial morphology, resembling the features of ferroptosis, a form of programmed cell death that is driven by iron-dependent accumulation of lipid peroxidation. Glutathione peroxidase 4 (GPX4) is the major lipid peroxidation scavenger and is necessary for TFH cell survival. The deletion of GPX4 in T cells selectively abrogated TFH cells and germinal center responses in immunized mice. Selenium supplementation enhanced GPX4 expression in T cells, increased TFH cell numbers and promoted antibody responses in immunized mice and young adults after influenza vaccination. Our findings reveal the central role of the selenium-GPX4-ferroptosis axis in regulating TFH homeostasis, which can be targeted to enhance TFH cell function in infection and following vaccination.


Asunto(s)
Ferroptosis/fisiología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Selenio/farmacología , Células T Auxiliares Foliculares/fisiología , Adolescente , Adulto , Animales , Supervivencia Celular/inmunología , Niño , Femenino , Centro Germinal/citología , Centro Germinal/inmunología , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Inmunidad Humoral/inmunología , Vacunas contra la Influenza/inmunología , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/fisiología , Ovalbúmina , Células T Auxiliares Foliculares/inmunología , Vacunación , Adulto Joven
7.
J Nanobiotechnology ; 19(1): 146, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011375

RESUMEN

BACKGROUND: Paclitaxel (PTX) has been suggested to be a promising front-line drug for gastric cancer (GC), while P-glycoprotein (P-gp) could lead to drug resistance by pumping PTX out of GC cells. Consequently, it might be a hopeful way to combat drug resistance by inhibiting the out-pumping function of P-gp. RESULTS: In this study, we developed a drug delivery system incorporating PTX onto polyethylene glycol (PEG)-modified and oxidized sodium alginate (OSA)-functionalized graphene oxide (GO) nanosheets (NSs), called PTX@GO-PEG-OSA. Owing to pH/thermal-sensitive drug release properties, PTX@GO-PEG-OSA could induced more obvious antitumor effects on GC, compared to free PTX. With near infrared (NIR)-irradiation, PTX@GO-PEG-OSA could generate excessive reactive oxygen species (ROS), attack mitochondrial respiratory chain complex enzyme, reduce adenosine-triphosphate (ATP) supplement for P-gp, and effectively inhibit P-gp's efflux pump function. Since that, PTX@GO-PEG-OSA achieved better therapeutic effect on PTX-resistant GC without evident toxicity. CONCLUSIONS: In conclusion, PTX@GO-PEG-OSA could serve as a desirable strategy to reverse PTX's resistance, combined with chemo/photothermal/photodynamic therapy.


Asunto(s)
Adenosina Trifosfato/metabolismo , Grafito/química , Grafito/farmacología , Mitocondrias/efectos de los fármacos , Paclitaxel/farmacología , Fotoquimioterapia/métodos , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Nanopartículas/química , Nanopartículas/uso terapéutico , Fototerapia , Polietilenglicoles , Células RAW 264.7 , Especies Reactivas de Oxígeno
8.
J Nanobiotechnology ; 18(1): 99, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32690085

RESUMEN

BACKGROUND: CuS-modified hollow mesoporous organosilica nanoparticles (HMON@CuS) have been preferred as non-invasive treatment for cancer, as near infrared (NIR)-induced photo-thermal effect (PTT) and/or photo-dynamic effect (PDT) could increase cancer cells' apoptosis. However, the certain role of HMON@CuS-produced-PTT&PDT inducing gastric cancer (GC) cells' mitochondrial damage, remained unclear. Moreover, theranostic efficiency of HMON@CuS might be well improved by applying multi-modal imaging, which could offer an optimal therapeutic region and time window. Herein, new nanotheranostics agents were reported by Gd doped HMON decorated by CuS nanocrystals (called HMON@CuS/Gd). RESULTS: HMON@CuS/Gd exhibited appropriate size distribution, good biocompatibility, L-Glutathione (GSH) responsive degradable properties, high photo-thermal conversion efficiency (82.4%) and a simultaneous reactive oxygen species (ROS) generation effect. Meanwhile, HMON@CuS/Gd could efficiently enter GC cells, induce combined mild PTT (43-45 °C) and PDT under mild NIR power density (0.8 W/cm2). Surprisingly, it was found that PTT might not be the only factor of cell apoptosis, as ROS induced by PDT also seemed playing an essential role. The NIR-induced ROS could attack mitochondrial transmembrane potentials (MTPs), then promote mitochondrial reactive oxygen species (mitoROS) production. Meanwhile, mitochondrial damage dramatically changed the expression of anti-apoptotic protein (Bcl-2) and pro-apoptotic protein (Bax). Since that, mitochondrial permeability transition pore (mPTP) was opened, followed by inducing more cytochrome c (Cyto C) releasing from mitochondria into cytosol, and finally activated caspase-9/caspase-3-depended cell apoptosis pathway. Our in vivo data also showed that HMON@CuS/Gd exhibited good fluorescence (FL) imaging (wrapping fluorescent agent), enhanced T1 imaging under magnetic resonance imaging (MRI) and infrared thermal (IRT) imaging capacities. Guided by FL/MRI/IRT trimodal imaging, HMON@CuS/Gd could selectively cause mild photo-therapy at cancer region, efficiently inhibit the growth of GC cells without evident systemic toxicity in vivo. CONCLUSION: HMON@CuS/Gd could serve as a promising multifunctional nanotheranostic platform and as a cancer photo-therapy agent through inducing mitochondrial dysfunction on GC.


Asunto(s)
Mitocondrias , Imagen Multimodal/métodos , Compuestos de Organosilicio , Fototerapia/métodos , Neoplasias Gástricas , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sulfato de Cobre , Humanos , Imagen por Resonancia Magnética , Mitocondrias/patología , Mitocondrias/efectos de la radiación , Compuestos de Organosilicio/química , Compuestos de Organosilicio/farmacología , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Nanomedicina Teranóstica
9.
Int Immunopharmacol ; 16(3): 376-81, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23474188

RESUMEN

Produced by CD4(+) helper T cells and natural killer T (NKT) cells, interleukin-21 (IL-21) performs broad regulatory functions on B cells, CD4(+) T cells, CD8(+) T cells, NK cells and NKT cells. Targeting IL-21 to enhance the immune system has attracted great interests in the development of vaccination, anti-infection and anti-tumor therapies. Administration of IL-21 in pre-clinical models is however limited by relatively high expense of the recombinant IL-21 protein. Here, we report a rapid and cost-effective method to produce IL-21 using Escherichia coli (E. coli) by introducing a novel two-step dilution strategy for refolding. The method has been validated to produce milligrams of human IL-21, human IL-21/IL-4 chimera and mouse IL-21 with high bioactivities and low endotoxin, mostly suitable for in vitro and in vivo pre-clinical studies.


Asunto(s)
Escherichia coli/metabolismo , Interleucinas/biosíntesis , Animales , Línea Celular , Evaluación Preclínica de Medicamentos , Escherichia coli/genética , Humanos , Subunidad alfa del Receptor de Interleucina-21/metabolismo , Interleucinas/química , Ratones , Pliegue de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA