Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cancer ; 15(10): 3128-3139, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706912

RESUMEN

Background: The long noncoding RNA small nucleolar RNA host gene 1 (SNHG1) has been demonstrated to play a crucial role in the progression of esophageal squamous cell carcinoma (ESCC). The current study aims to explore the deeper molecular mechanisms of SNHG1 in ESCC. Methods: Fifty patients with ESCC were enrolled to assess overall survival. Quantitative real-time PCR was performed to measure the levels of SNHG1, miR-216a-3p, and TMBIM6 in ESCC cells. Functional assessments of SNHG1 on ESCC cells were conducted using CCK-8 assay, flow cytometry, and Transwell assays. Western blot was conducted to detect the protein levels of TMBIM6 and proapoptotic proteins (Calpain and Caspase-12). The interaction among SNHG1, miR-216a-3p, and TMBIM6 was assessed with luciferase reporter assays. Results: Our study revealed that SNHG1 was notably increased in both clinical ESCC samples and cellular lines. Upregulation of SNHG1 in ESCC tissues was indicative of poor overall survival. Functionally, SNHG1 knockdown significantly inhibited the proliferation, migration, and invasion while promoting apoptosis in ESCC cells. Mechanistically, SNHG1 functioned as a competing endogenous RNA by sequestering miR-216a-3p to modulate TMBIM6 levels in ESCC cells. Notably, inhibiting miR-216a-3p or restoring TMBIM6 reversed the inhibitory effect induced by SNHG1 knockdown in ESCC cells. Conclusions: We demonstrate for the first time that SNHG1 may act as a competing endogenous RNA and promote ESCC progression through the miR-216a-3p/TMBIM6 axis. This highlights the potential of SNHG1 as a target for ESCC treatment.

2.
PLoS One ; 19(2): e0291543, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38354108

RESUMEN

Our previous work demonstrated that the anisodamine (ANI) and neostigmine (NEO) combination produced an antiseptic shock effect and rescued acute lethal crush syndrome by activating the α7 nicotinic acetylcholine receptor (α7nAChR). This study documents the therapeutic effect and underlying mechanisms of the ANI/NEO combination in dextran sulfate sodium (DSS)-induced colitis. Treating mice with ANI and NEO at a ratio of 500:1 alleviated the DSS-induced colitis symptoms, reduced body weight loss, improved the disease activity index, enhanced colon length, and alleviated colon inflammation. The combination treatment also enhanced autophagy in the colon of mice with DSS-induced colitis and lipopolysaccharide/DSS-stimulated Caco-2 cells. Besides, the ANI/NEO treatment significantly reduced INF-γ, TNF-α, IL-6, and IL-22 expression in colon tissues and decreased TNF-α, IL-1ß, and IL-6 mRNA levels in Caco-2 cells. Meanwhile, the autophagy inhibitor 3-methyladenine and ATG5 siRNA attenuated these effects. Furthermore, 3-methyladenine (3-MA) and the α7nAChR antagonist methyllycaconitine (MLA) weakened the ANI/NEO-induced protection on DSS-induced colitis in mice. Overall, these results indicate that the ANI/NEO combination exerts therapeutic effects through autophagy and α7nAChR in a DSS-induced colitis mouse model.


Asunto(s)
Colitis , Neostigmina , Alcaloides Solanáceos , Ratones , Animales , Humanos , Neostigmina/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Interleucina-6/metabolismo , Células CACO-2 , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Autofagia , Sulfato de Dextran/toxicidad , Colon/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
3.
EBioMedicine ; 90: 104507, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36893588

RESUMEN

BACKGROUND: Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease affecting multiple organs and tissues with high cellular heterogeneity. CD8+ T cell activity is involved in the SLE pathogenesis. However, the cellular heterogeneity and the underlying mechanisms of CD8+ T cells in SLE remain to be identified. METHODS: Single-cell RNA sequencing (scRNA-seq) of PBMCs from a SLE family pedigree (including 3 HCs and 2 SLE patients) was performed to identify the SLE-associated CD8+ T cell subsets. Flow cytometry analysis of a SLE cohort (including 23 HCs and 33 SLE patients), qPCR analysis of another SLE cohort (including 30 HCs and 25 SLE patients) and public scRNA-seq datasets of autoimmune diseases were employed to validate the finding. Whole-exome sequencing (WES) of this SLE family pedigree was used to investigate the genetic basis in dysregulation of CD8+ T cell subsets identified in this study. Co-culture experiments were performed to analyze the activity of CD8+ T cells. FINDINGS: We elucidated the cellular heterogeneity of SLE and identified a new highly cytotoxic CD8+ T cell subset, CD161-CD8+ TEMRA cell subpopulation, which was remarkably increased in SLE patients. Meanwhile, we discovered a close correlation between mutation of DTHD1 and the abnormal accumulation of CD161-CD8+ TEMRA cells in SLE. DTHD1 interacted with MYD88 to suppress its activity in T cells and DTHD1 mutation promoted MYD88-dependent pathway and subsequently increased the proliferation and cytotoxicity of CD161-CD8+ TEMRA cells. Furthermore, the differentially expressed genes in CD161-CD8+ TEMRA cells displayed a strong out-of-sample prediction for case-control status of SLE. INTERPRETATION: This study identified DTHD1-associated expansion of CD161-CD8+ TEMRA cell subpopulation is critical for SLE. Our study highlights genetic association and cellular heterogeneity of SLE pathogenesis and provides a mechanistical insight into the diagnosis and treatment of SLE. FUNDINGS: Stated in the Acknowledgements section of the manuscript.


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Humanos , Linfocitos T CD8-positivos , Factor 88 de Diferenciación Mieloide/metabolismo , Subgrupos de Linfocitos T , Linfocitos T Citotóxicos/metabolismo , Lupus Eritematoso Sistémico/genética , Enfermedades Autoinmunes/metabolismo
4.
Ann Transl Med ; 10(22): 1205, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36544639

RESUMEN

Background: Proprotein convertase subtilisin/kexin type 9 (PCSK9) belongs to an intracellular invertase or decarboxylase and is an independent risk factor for atherosclerosis (AS). This study aimed to investigate the therapeutic potential of the PCSK9 inhibitor, inclisiran, and its underlying mechanism in AS. Methods: ApoE-/- mice were fed with a high-fat diet (HFD) and intraperitoneally injected with 1, 5, or 10 mg/kg inclisiran. Low-density lipoprotein cholesterol (LDL-C), total cholesterol (TC), triglyceride (TG), and high-density lipoprotein cholesterol (HDL-C) levels were determined using commercially available kits. Oil Red O staining was applied to detect the aortic plaque area and oil formation. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to induce cell injuries. Cell death was determined using a Hoechst 33342/propidium iodide (PI) dual-staining assay. Cytotoxicity was measured by lactate dehydrogenase (LDH) activity analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analyses were performed to examine the pyroptosis-related factors. Results: Inclisiran inhibited the levels of LDL-C, TC, and TG, but increased the HDL-C level in the AS animal model. It also significantly inhibited plaque and oil droplet formation in a dose-dependent manner. Moreover, inclisiran markedly inhibited pyroptosis, as evidenced by the decreased levels of cleaved-caspase-1, NOD-like receptor family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-1 recruitment domain (ASC), gasdermin-D (GSDMD)-N, interleukin (IL)-1ß, and IL-18. Furthermore, inclisiran substantially inhibited cell death and cytotoxicity induced by ox-LDL in HUVECs. Conclusions: Inclisiran exerted an anti-atherosclerotic effect by inhibiting pyroptosis. This study provides a theoretical basis for the therapeutic potential of inclisiran in AS.

5.
Front Pharmacol ; 13: 933732, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36160450

RESUMEN

Ferritinophagy is a type of autophagy mediated by nuclear receptor activator 4 (NCOA4), which plays a role in inducing ferroptosis by regulating iron homeostasis and producing reactive oxygen species in cells. Under physiological conditions, ferritinophagy maintains the stability of intracellular iron by regulating the release of free iron. Studies have demonstrated that ferritinophagy is necessary to induce ferroptosis; however, under pathological conditions, excessive ferritinophagy results in the release of free iron in large quantities, which leads to lipid peroxidation and iron-dependent cell death, known as ferroptosis. Ferritinophagy has become an area of interest in recent years. We here in review the mechanism of ferritinophagy and its association with ferroptosis and various diseases to provide a reference for future clinical and scientific studies.

6.
Biochem Biophys Res Commun ; 616: 8-13, 2022 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-35636257

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) provoked a pandemic of acute respiratory disease, namely coronavirus disease 2019 (COVID-19). Currently, effective drugs for this disease are urgently warranted. Anisodamine is a traditional Chinese medicine that is predicted as a potential therapeutic drug for the treatment of COVID-19. Therefore, this study aimed to investigate its antiviral activity and crucial targets in SARS-CoV-2 infection. SARS-CoV-2 and anisodamine were co-cultured in Vero E6 cells, and the antiviral activity of anisodamine was assessed by immunofluorescence assay. The antiviral activity of anisodamine was further measured by pseudovirus entry assay in HEK293/hACE2 cells. Finally, the predictions of crucial targets of anisodamine on SARS-CoV-2 were analyzed by molecular docking studies. We discovered that anisodamine suppressed SARS-CoV-2 infection in Vero E6 cells, and reduced the SARS-CoV-2 pseudovirus entry to HEK293/hACE2 cells. Furthermore, molecular docking studies indicated that anisodamine may target SARS-CoV-2 main protease (Mpro) with the docking score of -6.63 kcal/mol and formed three H-bonds with Gly143, Cys145, and Cys44 amino acid residues at the predicted active site of Mpro. This study suggests that anisodamine is a potent antiviral agent for treating COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Proteasas 3C de Coronavirus , SARS-CoV-2 , Alcaloides Solanáceos , Antivirales/química , Antivirales/farmacología , COVID-19/virología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/efectos de los fármacos , Proteasas 3C de Coronavirus/metabolismo , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Péptido Hidrolasas , Inhibidores de Proteasas/farmacología , Alcaloides Solanáceos/farmacología , Proteínas no Estructurales Virales/química
7.
Ann Transl Med ; 9(19): 1503, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34805365

RESUMEN

OBJECTIVE: We aimed at comprehensively analyzing ferroptosis regulation and its potential role in the treatment of associated diseases. BACKGROUND: Ferroptosis is a recently discovered form of cell death that involves small molecule-induced oxidative cell death. This process is usually accompanied by large amounts of iron accumulation and lipid peroxidation. Ferroptosis inducers directly or indirectly affect glutathione peroxidase (GPXs) through different pathways. Disturbances in GPXs result in suppressed cellular antioxidant capacities, accumulation of lipid reactive oxygen species (ROS) and oxidative cell death. It has been reported that ferroptosis is closely associated with the pathophysiological processes of many diseases, including tumors, nervous system diseases, ischemia-reperfusion injury, kidney injury and iron metabolism diseases among others. METHODS: First, we reviewed the mechanisms of ferroptosis, with emphasis on the characteristics and functions of ferroptosis in multiple pathways. Then, inducers and inhibitors of ferroptosis were reviewed, and their mechanisms of action elucidated. Finally, ferroptosis-associated pathophysiological processes of various diseases were reviewed. CONCLUSIONS: Ferroptosis is associated with the occurrence and development of various diseases. Elucidation of the mechanisms involved in ferroptosis will inform new therapeutic targets and strategies for these diseases.

8.
Front Pharmacol ; 12: 676589, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995108

RESUMEN

The α7 nicotinic acetylcholine receptor (α7nAChR) belongs to the superfamily of cys loop cationic ligand-gated channels, which consists of homogeneous α7 subunits. Although our lab found that activation of α7nAChR could alleviate ischemic stroke, the mechanism is still unknown. Herein, we explored whether autophagy is involved in the neuroprotective effect mediated by α7nAChR in ischemic stroke. Transient middle cerebral artery occlusion (tMCAO) and oxygen and glucose deprivation (OGD/R) exposure were applied to in vivo and in vitro models of ischemic stroke, respectively. Neurological deficit score and infarct volume were used to evaluate outcomes of tMCAO in the in vivo study. Autophagy-related proteins were detected by Western blot, and autophagy flux was detected by using tandem fluorescent mRFP-GFP-LC3 lentivirus. At 24 h after tMCAO, α7nAChR knockout mice showed worse neurological function and larger infarct volume than wild-type mice. PNU282987, an α7nAChR agonist, protected against OGD/R-induced neuronal injury, enhanced autophagy, and promoted autophagy flux. However, the beneficial effects of PNU282987 were eliminated by 3-methyladenine (3-MA), an autophagy inhibitor. Moreover, we found that PNU282987 treatment could activate the AMPK-mTOR-p70S6K signaling pathway in the in vitro study, while the effect was attenuated by compound C, an AMPK inhibitor. Our results demonstrated that the beneficial effect on neuronal survival via activation of α7nAChR was associated with enhanced autophagy, and the AMPK-mTOR-p70S6K signaling pathway was involved in α7nAChR activation-mediated neuroprotection.

9.
J Mater Chem B ; 3(39): 7787-7795, 2015 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-32264587

RESUMEN

A human umbilical vein endothelial cell/human dermal fibroblast (HUVEC/HDF) co-culture system has been widely applied for mimicking the vascularization process in tissue engineering. Here, we have developed plasmonic superparamagnetic nanoshells (SNs) to realize the visualization of cell proliferation behaviour via two-photon luminescence of particle shells and the cell separation via the superparamagnetic properties of particle cores. The cell viability test and gene expression analysis have demonstrated no obvious cytotoxicity and disturbance to cell co-culture. Cell separation efficiency via SNs reaches a value of 94.7%, close to 99.5% in routine methods by magnetic beads. In contrast to the complicated and expensive process using specific antibody-targeted immunofluorescence staining and the magnetic beads separation in routine methods, SNs present a more simple and effective way to achieve both functions with better photo-stability and a negligible photo-bleaching effect via TPL imaging. Furthermore, the combination of the TPL imaging and magnetic manipulation of SNs also offers the potential of spontaneously enhancing the cell seeding and tracking the cell distribution in 3D tissue engineering scaffolds.

10.
Nanoscale ; 6(23): 14360-70, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25329447

RESUMEN

With the increasing need for multi-purpose analysis in the biomedical field, traditional single diagnosis methods cannot meet the requirements. Therefore new multifunctional technologies and materials for the integration of sample collection, sensing and imaging are in great demand. Core-shell nanoparticles offer a unique platform to combine multifunctions in a single particle. In this work, we have constructed a novel type of core-shell superparamagnetic nanoshell (Fe3O4@SiO2@Au), composed of a Fe3O4 cluster core, a thin Au shell and a SiO2 layer in between. The obtained multifunctional nanoparticles combine the magnetic properties and plasmonic optical properties effectively, which were well investigated by a number of experimental characterization methods and theoretical simulations. We have demonstrated that Fe3O4@SiO2@Au nanoparticles can be utilized for two-photon luminescence (TPL) imaging, near-infrared surface-enhanced Raman scattering (NIR SERS) and cell collection by magnetic separation. The TPL intensity could be further greatly enhanced through the plasmon coupling effect in the self-assembled nanoparticle chains, which were triggered by an external magnetic field. In addition, Fe3O4@SiO2@Au nanoparticles may have great potential applications such as enhanced magnetic resonance imaging (MRI) and photo-thermotherapy. Successful combination of multifunctions including magnetic response, biosensing and bioimaging in single nanoparticles allows further manipulation, real-time tracking, and intracellular molecule analysis of live cells at a single-cell level.


Asunto(s)
Óxido Ferrosoférrico , Oro , Células Endoteliales de la Vena Umbilical Humana/citología , Luminiscencia , Nanocáscaras/química , Imagen Óptica/métodos , Dióxido de Silicio , Óxido Ferrosoférrico/química , Óxido Ferrosoférrico/farmacología , Oro/química , Oro/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Imagen por Resonancia Magnética/métodos , Dióxido de Silicio/química , Dióxido de Silicio/farmacología
11.
Biomaterials ; 35(12): 3803-18, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24486216

RESUMEN

The facts that biomaterials affect the behavior of single type of cells have been widely accepted. However, the effects of biomaterials on cell-cell interactions have rarely been reported. Bone tissue engineering involves osteoblastic cells (OCs), endothelial cells (ECs) and the interactions between OCs and ECs. It has been reported that silicate biomaterials can stimulate osteogenic differentiation of OCs and vascularization of ECs. However, the effects of silicate biomaterials on the interactions between ECs and OCs during vascularization and osteogenesis have not been reported, which are critical for bone tissue regeneration in vivo. Therefore, this study aimed to investigate the effects of calcium silicate (CS) bioceramics on interactions between human umbilical vein endothelial cells (HUVECs) and human bone marrow stromal cells (HBMSCs) and on stimulation of vascularization and osteogenesis in vivo through combining co-cultures with CS containing scaffolds. Specifically, the effects of CS on the angiogenic growth factor VEGF, osteogenic growth factor BMP-2 and the cross-talks between VEGF and BMP-2 in the co-culture system were elucidated. Results showed that CS stimulated co-cultured HBMSCs (co-HBMSCs) to express VEGF and the VEGF activated its receptor KDR on co-cultured HUVECs (co-HUVECs), which was also up-regulated by CS. Then, BMP-2 and nitric oxide expression from the co-HUVECs were stimulated by CS and the former stimulated osteogenic differentiation of co-HBMSCs while the latter stimulated vascularization of co-HVUECs. Finally, the poly(lactic-co-glycolic acid)/CS composite scaffolds with the co-cultured HBMSCs and HUVECs significantly enhanced vascularization and osteogenic differentiation in vitro and in vivo, which indicates that it is a promising way to enhance bone regeneration by combining scaffolds containing silicate bioceramics and co-cultures of ECs and OCs.


Asunto(s)
Cerámica , Células Endoteliales/citología , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Osteogénesis , Silicatos/química , Secuencia de Bases , Proteína Morfogenética Ósea 2/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Cartilla de ADN , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor A de Crecimiento Endotelial Vascular/fisiología
12.
J Mater Sci Mater Med ; 25(3): 791-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24338379

RESUMEN

Fluoride treatment is a commonly used technique or pre-treatment to optimize the degradation kinetic and improve the biocompatibility of magnesium-based implant. The influence of changed surface properties and degradation kinetics on subsequent protein adsorption and cytocompatibility is critical to understand the biocompatibility of the implant. In this study, a patent magnesium alloy Mg-Nd-Zn-Zr alloy (JDBM) designed for cardiovascular stent application was treated by immersion in hydrofluoric acid. A 1.5 µm thick MgF2 layer was prepared. The surface roughness was increased slightly while the surface zeta potential was changed to a much more negative value after the treatment. Static contact angle test was performed, showing an increase in hydrophilicity and surface energy after the treatment. The MgF2 layer slowed down in vitro degradation rate, but lost the protection effect after 10 days. The treatment enhanced human albumin adsorption while no difference of human fibrinogen adsorption amount was observed. Direct cell adhesion test showed many more live HUVECs retained than bare magnesium alloy. Both treated and untreated JDBM showed no adverse effect on HUVEC viability and spreading morphology. The relationship between changed surface characteristics, degradation rate and protein adsorption, cytocompatibility was also discussed.


Asunto(s)
Materiales Biocompatibles/farmacología , Líquidos Corporales/química , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Fluoruros/química , Compuestos de Magnesio/síntesis química , Compuestos de Magnesio/farmacología , Aleaciones/síntesis química , Aleaciones/farmacología , Materiales Biocompatibles/síntesis química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Humanos , Ensayo de Materiales , Propiedades de Superficie
13.
J Mater Chem B ; 2(8): 1100-1110, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-32261627

RESUMEN

Copper (Cu) has been reported to be able to stimulate vascularization/angiogenesis, which is critical for regeneration of vascularized tissue in tissue engineering. Silicate bioceramics have also been reported to have stimulatory effects on vascularization due to the silicon (Si) ions released from silicate biomaterials. Therefore, we hypothesize that a combination of Cu and Si ions may show synergy effects on vascularization. Therefore, a copper-doped calcium silicate bioceramic (Cu-CaSiO3, Cu-CS) was designed and synthesized with the purpose to enhance the stimulatory effects of copper salts or pure silicate bioceramics on vascularization by combining the effects of Cu and Si ions. The cytocompatibility of Cu-CS was firstly assessed by testing the influence of Cu-CS ion extracts on proliferation of human umbilical vein endothelial cells (HUVECs). Thereafter, vascularization of HUVECs on ECMatrix™ gel or co-cultured with human dermal fibroblasts (HDFs) in Cu-CS extracts was evaluated and expression of angiogenic growth factors was analyzed. Results revealed that, as compared to CS extracts and media containing soluble CuSO4, Cu-CS extracts possessed stronger stimulatory effects on upregulation of angiogenic growth factors, which finally resulted in better stimulatory effects on vascularization. During the vascularization process, paracrine effects dominated in the co-culture system. In addition, lower concentrations of Cu and Si ions released from Cu-CS than those released from pure CS or CuSO4 were enough to stimulate vascularization, which indicated that there were synergy effects between Cu and Si ions during stimulation of vascularization by Cu-CS. Taken together, the designed Cu-CS may be suitable as a new biomaterial for regenerating blood vessels in tissue engineering.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA