Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 711: 149920, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38615574

RESUMEN

Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.


Asunto(s)
Antituberculosos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Mycobacterium tuberculosis , Transducción de Señal , Tuberculosis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/inmunología , Transducción de Señal/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Tuberculosis/inmunología , Tuberculosis/metabolismo , Tuberculosis/microbiología , Animales , Antituberculosos/uso terapéutico , Antituberculosos/farmacología , Terapia Molecular Dirigida/métodos
2.
Molecules ; 29(5)2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38474476

RESUMEN

Major Depressive Disorder (MDD) is a complex mental disorder that involves alterations in signal transmission across multiple scales and structural abnormalities. The development of effective antidepressants (ADs) has been hindered by the dominance of monoamine hypothesis, resulting in slow progress. Traditional ADs have undesirable traits like delayed onset of action, limited efficacy, and severe side effects. Recently, two categories of fast-acting antidepressant compounds have surfaced, dissociative anesthetics S-ketamine and its metabolites, as well as psychedelics such as lysergic acid diethylamide (LSD). This has led to structural research and drug development of the receptors that they target. This review provides breakthroughs and achievements in the structure of depression-related receptors and novel ADs based on these. Cryo-electron microscopy (cryo-EM) has enabled researchers to identify the structures of membrane receptors, including the N-methyl-D-aspartate receptor (NMDAR) and the 5-hydroxytryptamine 2A (5-HT2A) receptor. These high-resolution structures can be used for the development of novel ADs using virtual drug screening (VDS). Moreover, the unique antidepressant effects of 5-HT1A receptors in various brain regions, and the pivotal roles of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) and tyrosine kinase receptor 2 (TrkB) in regulating synaptic plasticity, emphasize their potential as therapeutic targets. Using structural information, a series of highly selective ADs were designed based on the different role of receptors in MDD. These molecules have the favorable characteristics of rapid onset and low adverse drug reactions. This review offers researchers guidance and a methodological framework for the structure-based design of ADs.


Asunto(s)
Trastorno Depresivo Mayor , Humanos , Trastorno Depresivo Mayor/tratamiento farmacológico , Serotonina , Estructura Molecular , Microscopía por Crioelectrón , Antidepresivos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
3.
J Nanobiotechnology ; 21(1): 369, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37817142

RESUMEN

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, is still one of the top killers worldwide among infectious diseases. The escape of Mtb from immunological clearance and the low targeting effects of anti-TB drugs remain the substantial challenges for TB control. Iron is particularly required for Mtb growth but also toxic for Mtb in high dosages, which makes iron an ideal toxic decoy for the 'iron-tropic' Mtb. Here, a macrophage-targeted iron oxide nanoparticles (IONPs)-derived IONPs-PAA-PEG-MAN nanodecoy is designed to augment innate immunological and drug killings against intracellular Mtb. IONPs-PAA-PEG-MAN nanodecoy exhibits preferential uptake in macrophages to significantly increase drug uptake with sustained high drug contents in host cells. Moreover, it can serve as a specific nanodecoy for the 'iron-tropic' Mtb to realize the localization of Mtb contained phagosomes surrounding the drug encapsulated nanodecoys and co-localization of Mtb with the drug encapsulated nanodecoys in lysosomes, where the incorporated rifampicin (Rif) can be readily released under acidic lysosomal condition for enhanced Mtb killing. This drug encapsulated nanodecoy can also polarize Mtb infected macrophages into anti-mycobacterial M1 phenotype and enhance M1 macrophage associated pro-inflammatory cytokine (TNF-α) production to trigger innate immunological responses against Mtb. Collectively, Rif@IONPs-PAA-PEG-MAN nanodecoy can synergistically enhance the killing efficiency of intracellular Mtb in in vitro macrophages and ex vivo monocyte-derived macrophages, and also significantly reduce the mycobacterial burdens in the lung of infected mice with alleviated pathology. These results indicate that Rif@IONPs-PAA-PEG-MAN nanodecoy may have a potential for the development of more effective therapeutic strategy against TB by manipulating augmented innate immunity and drug killings.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Humanos , Animales , Ratones , Macrófagos , Tuberculosis/tratamiento farmacológico , Rifampin/farmacología , Hierro
4.
Curr Microbiol ; 80(5): 171, 2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37024713

RESUMEN

Dengue remains a public health issue worldwide. Similar to chronic infectious diseases, stimulation of cytokine production is not enough to drive immune effector cells for effective virus clearance. One possible mechanism is the virus induces a large number of negative stimulatory cytokines inhibiting immune response. Interleukin 37 (IL-37) plays a crucial regulatory role in infection and immunity, inhibits innate and adaptive immunity as an anti-inflammatory cytokine by inhibiting proinflammatory mediators and pathways. To date, there are few studies reporting correlations between dengue fever (DF) and IL-37. In this study we found that the serum IL-37b and IL-37b-producing monocytes in patients were significantly increased in DF patients. A majority of the IL-37b produced by DF patients was produced by monocytes, not lymphocytes. Increased levels of IL-6, IL-10, and IFN-α were also found in DF patients. However, we failed to detect IL-1ß, IL-17A and TNF-α in plasma, because of off-target. In our study, there was no relation between IL-6, IL-10, and IFN-α expressions and IL-37b in serum (P > 0.05). The IL-37b-producing monocytes were negatively correlated with the level of IFN-α in serum and platelet count, and positively correlated with lymphocytes percentage (P < 0.05, respectively). Additionally, serum DENV nonstructural protein 1 levels were positively correlated with monocytes percentages (P < 0.05). Our data represents findings for IL-37b expression and its potential mechanisms in DF patients' immune response.


Asunto(s)
Virus del Dengue , Dengue , Humanos , Interleucina-10 , Virus del Dengue/fisiología , Interleucina-6 , Carga Viral , Citocinas
5.
Int J Med Microbiol ; 312(7): 151569, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36274382

RESUMEN

Tuberculosis (TB) induced by Mycobacterium tuberculosis (M. tuberculosis) infection remains a global most deadly infectious disease. While development of more effective TB vaccines and therapeutics relies on identifications of true biomarkers designating an immune protection against M. tuberculosis infection, exact protective immune components against M. tuberculosis infection remain largely unidentified. We previously found that severe TB induced remarkable up-regulation of interferon regulatory factor 7 (IRF7) and IRF7-related gene signatures, implicating that some unknown downstream molecules in IRF7 signaling cascades may determine the M. tuberculosis infection outcomes and serve as a protective immune component against M. tuberculosis infection. Indeed, here, we observe that genetic ablation of IRF7 leads to more severe lung pathology, increased M. tuberculosis burdens, impaired differentiation of effector/memory T subsets, and extensively elevated expression of pro-inflammatory cytokines in lungs. Importantly, IRF7 is vital for sustaining expression of PD-1/PD-L1 and PD-1/PD-L1-modulated miRNA-31. Moreover, interventions of miRNA-31 expressions via administration of miRNA-31 agomir reduces lung pathology and bacilli burdens via inducing up-regulation of gene sets involved in biological processes of defense response or cellular and chemical homeostasis in lungs. Thus, this study uncovers previously unrecognized importance and mechanisms of IRF7-mediated miRNA-31 as a protective immune component against M. tuberculosis infection.


Asunto(s)
MicroARNs , Mycobacterium tuberculosis , Tuberculosis , Humanos , Antígeno B7-H1 , Factor 7 Regulador del Interferón/genética , Receptor de Muerte Celular Programada 1 , Tuberculosis/microbiología , MicroARNs/genética
6.
J Nanobiotechnology ; 20(1): 36, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35033108

RESUMEN

Tuberculosis (TB), induced by Mycobacterium tuberculosis (Mtb) infection, remains a top killer among infectious diseases. While Bacillus Calmette-Guerin (BCG) is the sole TB vaccine, the clumped-clustered features of BCG in intradermal immunization appear to limit both the BCG protection efficacy and the BCG vaccination safety. We hypothesize that engineering of clumped-clustered BCG into nanoscale particles would improve safety and also facilitate the antigen-presenting-cell (APC)'s uptake and the following processing/presentation for better anti-TB protective immunity. Here, we engineered BCG protoplasts into nanoscale membraned BCG particles, termed as "BCG-Nanocage" to enhance the anti-TB vaccination efficiency and safety. BCG-Nanocage could readily be ingested/taken by APC macrophages selectively; BCG-Nanocage-ingested macrophages exhibited better viability and developed similar antimicrobial responses with BCG-infected macrophages. BCG-Nanocage, like live BCG bacilli, exhibited the robust capability to activate and expand innate-like T effector cell populations of Vγ2+ T, CD4+ T and CD8+ T cells of rhesus macaques in the ex vivo PBMC culture. BCG-Nanocage immunization of rhesus macaques elicited similar or stronger memory-like immune responses of Vγ2Vδ2 T cells, as well as Vγ2Vδ2 T and CD4+/CD8+ T effectors compared to live BCG vaccination. BCG-Nanocage- immunized macaques developed rapidly-sustained pulmonary responses of Vγ2Vδ2 T cells upon Mtb challenge. Furthermore, BCG- and BCG-Nanocage- immunized macaques, but not saline controls, exhibited undetectable Mtb infection loads or TB lesions in the Mtb-challenged lung lobe and hilar lymph node at endpoint after challenge. Thus, the current study well justifies a large pre-clinical investigation to assess BCG-Nanocage for safe and efficacious anti-TB vaccination, which is expected to further develop novel vaccines or adjuvants.


Asunto(s)
Vacuna BCG , Linfocitos T CD8-positivos/inmunología , Mycobacterium tuberculosis/inmunología , Nanoestructuras/química , Tuberculosis/inmunología , Animales , Vacuna BCG/química , Vacuna BCG/inmunología , Células Cultivadas , Femenino , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Macaca mulatta , Masculino
7.
Anal Bioanal Chem ; 413(30): 7401-7410, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34673992

RESUMEN

The resistance of urinary tract pathogenic bacteria to various antibiotics is increasing, which requires the rapid detection of infectious pathogens for accurate and timely antibiotic treatment. Here, we propose a rapid diagnosis strategy for the antibiotic resistance of bacteria in urinary tract infections (UTIs) based on surface-enhanced Raman scattering (SERS) using a positively charged gold nanoparticle planar solid SERS substrate. Then, an intelligent identification model for SERS spectra based on the deep learning technique is constructed to realize the rapid, ultrasensitive, and non-labeled detection of pathogenic bacteria. A total of 54,000 SERS spectra were collected from 18 isolates belonging to 6 species of common UTI bacteria in this work to realize identification of bacterial species, antibiotic sensitivity, and multidrug resistance (MDR) via convolutional neural networks (CNN). This method significantly simplify the Raman data processing processes without background removing and smoothing, however, achieving 96% above classification accuracy, which was significantly greater than the 85% accuracy of the traditional multivariate statistical analysis algorithm principal component analysis combined with the K-nearest neighbor (PCA-KNN). This work clearly elucidated the potential of combining SERS and deep learning technique to realize culture-free identification of pathogenic bacteria and their associated antibiotic sensitivity.


Asunto(s)
Aprendizaje Profundo , Farmacorresistencia Bacteriana , Espectrometría Raman/métodos , Infecciones Urinarias/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias/clasificación , Bacterias/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Especificidad de la Especie , Infecciones Urinarias/tratamiento farmacológico
8.
Angew Chem Int Ed Engl ; 59(8): 3226-3234, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31756258

RESUMEN

Pathogenesis hallmarks for tuberculosis (TB) are the Mycobacterium tuberculosis (Mtb) escape from phagolysosomal destruction and limited drug delivery into infected cells. Several nanomaterials can be entrapped in lysosomes, but the development of functional nanomaterials to promote phagolysosomal Mtb clearance remains a big challenge. Here, we report on the bactericidal effects of selenium nanoparticles (Se NPs) against Mtb and further introduce a novel nanomaterial-assisted anti-TB strategy manipulating Ison@Man-Se NPs for synergistic drug-induced and phagolysosomal destruction of Mtb. Ison@Man-Se NPs preferentially entered macrophages and accumulated in lysosomes releasing Isoniazid. Surprisingly, Ison@Man-Se/Man-Se NPs further promoted the fusion of Mtb into lysosomes for synergistic lysosomal and Isoniazid destruction of Mtb. Concurrently, Ison@Man-Se/Man-Se NPs also induced autophagy sequestration of Mtb, evolving into lysosome-associated autophagosomal Mtb degradation linked to ROS-mitochondrial and PI3K/Akt/mTOR signaling pathways. This novel nanomaterial-assisted anti-TB strategy manipulating antimicrobial immunity and Mtb clearance may potentially serve in more effective therapeutics against TB and drug-resistant TB.


Asunto(s)
Antibacterianos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Isoniazida/química , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Nanopartículas/química , Selenio/química , Tuberculosis/tratamiento farmacológico , Humanos , Tuberculosis/patología
9.
J Cell Biochem ; 120(3): 3736-3746, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30229997

RESUMEN

Oridonin, an active diterpenoid isolated from Rabdosia rubescens, has been reported for its antitumor activity on several cancers. However, its effect on human esophageal cancer remains unclear. In this study, we demonstrated that oridonin could inhibit the growth of human esophageal cancer cells both in vitro and in vivo. Oridonin not only suppressed the proliferation, but also induced cell cycle arrest and mitochondrial-mediated apoptosis in KYSE-30, KYSE-150, and EC9706 cells with dose-dependent manner. Further mechanism studies revealed that oridonin led cell cycle arrest in esophageal cancer cells via downregulating cell cycle-related proteins, such as cyclin B1 and CDK2, while upregulating p53 and p21. Oridonin also increased proapoptotic protein Bax and reduced antiapoptotic protein Bcl-2, as well as the increased expression of cleaved caspase-3, -8, and -9. In addition, oridonin treatment could significantly inhibit the PI3K/Akt/mTOR and Ras/Raf signaling pathway. In vivo results further demonstrated that oridonin treatment markedly inhibited tumor growth in the esophageal cancer xenograft mice model. Taken together, these results suggest that oridonin may be a potential anticancer agent for the treatment of esophageal cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Diterpenos de Tipo Kaurano/farmacología , Neoplasias Esofágicas/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina B1/antagonistas & inhibidores , Ciclina B1/genética , Ciclina B1/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/agonistas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Humanos , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/agonistas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/agonistas , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
10.
Respir Res ; 19(1): 199, 2018 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305102

RESUMEN

BACKGROUND: This study aimed at predicting the survival status on non-small cell lung cancer patients with the phenotypic radiomics features obtained from the CT images. METHODS: A total of 186 patients' CT images were used for feature extraction via Pyradiomics. The minority group was balanced via SMOTE method. The final dataset was randomized into training set (n = 223) and validation set (n = 75) with the ratio of 3:1. Multiple random forest models were trained applying hyperparameters grid search with 10-fold cross-validation using precision or recall as evaluation standard. Then a decision threshold was searched on the selected model. The final model was evaluated through ROC curve and prediction accuracy. RESULTS: From those segmented images of 186 patients, 1218 features were obtained via feature extraction. The preferred model was selected with recall as evaluation standard and the optimal decision threshold was set 0.56. The model had a prediction accuracy of 89.33% and the AUC score was 0.9296. CONCLUSION: A hyperparameters tuning random forest classifier had greater performance in predicting the survival status of non-small cell lung cancer patients, which could be taken for an automated classifier promising to stratify patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/mortalidad , Tomografía Computarizada por Rayos X/tendencias , Biomarcadores de Tumor , Bases de Datos Factuales/tendencias , Femenino , Humanos , Masculino , Valor Predictivo de las Pruebas , Tasa de Supervivencia/tendencias , Tomografía Computarizada por Rayos X/métodos
11.
Pharmacol Res ; 119: 479-489, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28411855

RESUMEN

As the active anticancer component of Rabdosia Rubescens, oridonin has been proved to show strong anticancer activity in cancer cells, which is also found to be closely related to its specific inhibition effects on the EGFR tyrosine kinase activity. In this study, atomic force microscopy based single molecule force spectroscopy (AFM-SMFS) was used for real-time and in-situ detection of EGF-EGFR interactions in living esophageal cancer KYSE-150 cells to evaluate the anticancer activity of oridonin for the first time. Oridonin was found to induce apoptosis and also reduce EGFR expression in KYSE-150 cells. AFM-SMFS results demonstrated that oridonin could inhibit the binding between EGF and EGFR in KYSE-150 cells by decreasing the unbinding force and binding probability for EGF-EGFR complexes, which was further proved to be closely associated with the intracellular ROS level. More precise mechanism studies based on AFM-SMFS demonstrated that oridonin treatment could decrease the energy barrier width, increase the dissociation off rate constant and decrease the activation energy of EGF-EGFR complexes in ROS dependent way, suggesting oridonin as a strong anticancer agent targeting EGF-EGFR interactions in cancer cells through ROS dependent mechanism. Our results not only suggested oridonin as a strong anticancer agent targeting EGF-EGFR interactions in ROS dependent mechanism, but also highlighted AFM-SMFS as a powerful technique for pharmacodynamic studies by detecting ligand-receptor interactions, which was also expected to be developed into a promising tool for the screening and mechanism studies of drugs.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Diterpenos de Tipo Kaurano/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Diterpenos de Tipo Kaurano/química , Neoplasias Esofágicas/metabolismo , Esófago/efectos de los fármacos , Esófago/metabolismo , Humanos , Isodon/química , Microscopía de Fuerza Atómica
12.
Pharmacol Res ; 111: 374-383, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27339828

RESUMEN

The degranulation of mast cells and basophils is often initiated by a number of pathophysiological responses, especially in allergic and inflammatory conditions. Efficient techniques and methods for determining the level of such degranulation are highly demanded for laboratory and clinical studies. In this work, a rapid and sensitive assay based on the particle analysis of granules in RBL-2H3 cells, a cell line widely used as a convenient model system to study the degranulation of mast cells and basophils, was developed to detect cell degranulation using a Nanosight NS300 in light scatter mode and dynamic light scattering (DLS) on a Malvern Zetasizer Nano-ZS instrument. Using this method, drug-induced mast cell degranulation and systemic anaphylaxis were efficiently determined both in cell culture medium and blood samples from animals in the current study. This promising method is expected to be widely used for screening anti-allergic and anti-inflammatory drugs both in vitro and in vivo models, as well as for determining the level of mast cell degranulation of the patients in the clinic.


Asunto(s)
Prueba de Desgranulación de los Basófilos/métodos , Basófilos/metabolismo , Técnicas Biosensibles , Degranulación de la Célula , Mastocitos/metabolismo , Animales , Prueba de Desgranulación de los Basófilos/instrumentación , Basófilos/efectos de los fármacos , Basófilos/inmunología , Calcio/metabolismo , Degranulación de la Célula/efectos de los fármacos , Línea Celular Tumoral , Dispersión Dinámica de Luz , Diseño de Equipo , Exocitosis/efectos de los fármacos , Femenino , Ionomicina/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Morfinanos/farmacología , Valor Predictivo de las Pruebas , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Tiempo , p-Metoxi-N-metilfenetilamina/farmacología
13.
Bioorg Med Chem Lett ; 26(11): 2730-4, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27080177

RESUMEN

According to the previous studies, the anticancer activity of flavonoids could be enhanced when they are coordinated with transition metal ions. In this work, kaempferol-zinc(II) complex (kaempferol-Zn) was synthesized and its chemical properties were characterized by UV-VIS, FT-IR, (1)H NMR, elemental analysis, electrospray mass spectrometry (ES-MS) and fluorescence spectroscopy, which showed that the synthesized complex was coordinated with a Zn(II) ion via the 3-OH and 4-oxo groups. The anticancer effects of kaempferol-Zn and free kaempferol on human oesophageal cancer cell line (EC9706) were compared. MTT results demonstrated that the killing effect of kaempferol-Zn was two times higher than that of free kaempferol. Atomic force microscopy (AFM) showed the morphological and ultrastructural changes of cellular membrane induced by kaempferol-Zn at subcellular or nanometer level. Moreover, flow cytometric analysis indicated that kaempferol-Zn could induce apoptosis in EC9706 cells by regulating intracellular calcium ions. Collectively, all the data showed that kaempferol-Zn might be served as a kind of potential anticancer agent.


Asunto(s)
Antineoplásicos/farmacología , Quempferoles/farmacología , Compuestos Organometálicos/farmacología , Zinc/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Quempferoles/química , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Espectrometría de Fluorescencia , Relación Estructura-Actividad , Zinc/química
14.
Anal Bioanal Chem ; 408(1): 165-76, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26476923

RESUMEN

A new method based on atomic force microscopy (AFM) was developed to investigate the anti-inflammatory effects of drugs on lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. The LPS-stimulated RAW264.7 macrophage cell line is a widely used in vitro cell model for the screening of anti-inflammatory drugs or the study of anti-inflammatory mechanisms. In this work, the inhibitory effects of dexamethasone and quercetin on LPS-CD14 receptor binding in RAW264.7 macrophages was probed by LPS-functionalized tips for the first time. Both dexamethasone and quercetin were found to inhibit LPS-induced NO production, iNOS expression, IκBα phosphorylation, and IKKα/ß phosphorylation in RAW264.7 macrophages. The morphology and ultrastructure of RAW264.7 macrophages were determined by AFM, which indicated that dexamethasone and quercetin could inhibit LPS-induced cell surface particle size and roughness increase in RAW264.7 macrophages. The binding of LPS and its receptor in RAW264.7 macrophages was determined by LPS-functionalized AFM tips, which demonstrated that the binding force and binding probability between LPS and CD14 receptor on the surface of RAW264.7 macrophages were also inhibited by dexamethasone or quercetin treatment. The obtained results imply that AFM, which is very useful for the investigation of potential targets for anti-inflammatory drugs on native macrophages and the enhancement of our understanding of the anti-inflammatory effects of drugs, is expected to be developed into a promising tool for the study of anti-inflammatory drugs.


Asunto(s)
Antiinflamatorios/farmacología , Dexametasona/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Microscopía de Fuerza Atómica/métodos , Quercetina/farmacología , Animales , Lipopolisacáridos/inmunología , Macrófagos/química , Ratones , FN-kappa B/inmunología , Óxido Nítrico/inmunología , Células RAW 264.7
15.
Appl Microbiol Biotechnol ; 100(15): 6643-6652, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26883344

RESUMEN

Angiogenesis provides necessary nutrients and oxygen for tumor growth and metastasis; thus, every stage of angiogenesis process is the potential target for cancer therapies. Ursolic acid (UA) is reported to decrease tumor burden through anti-angiogenesis pathway, but its poor water solubility greatly limits its efficiency and clinical application. Here, a simple method for preparing UA-loaded chitosan nanoparticles (CH-UA-NPs) with anti-angiogenesis and anti-tumor activity was demonstrated. In vitro, CH-UA-NPs could significantly inhibit the proliferation, migration, and tube formation of human umbilical vascular endothelial cells (HUVECs). After uptake by HUVECs, CH-UA-NPs were mainly localized in lysosomes and mitochondria, but not nuclei. CH-UA-NPs induced the destruction of lysosome membrane integrity, collapse of mitochondrial membrane potential, and reorganization of cell cytoskeleton. All these changes led to the apoptosis or necrosis in HUVECs. In vivo, CH-UA-NPs could inhibit the angiogenesis in chicken chorioallantoic membrane (CAM) model and H22 xenograft model. Notably, comparing with free UA, such synthesized CH-UA-NPs could save about tenfold of UA doses, implying that this could significantly decrease the side effects induced by high doses of UA in biological organism. Our data showed that CH-UA-NPs and this nanoparticle-based drug delivery system could be as a potential drug candidate for anti-angiogenesis treatment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Quitosano/química , Membrana Corioalantoides/irrigación sanguínea , Portadores de Fármacos/química , Nanopartículas/química , Neovascularización Patológica/prevención & control , Triterpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Triterpenos/química , Cicatrización de Heridas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Ursólico
16.
Anal Chem ; 87(9): 4797-805, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25851113

RESUMEN

Quartz crystal microbalance with dissipation (QCM-D) monitoring was used for real-time and label-free detection of changes and folate receptor (FR) expression levels on living MCF-7 cells for evaluating the anticancer activity of resveratrol. Here, the mechanical changes of cellular responses to resveratrol were tracked by a poly(l-lysine) (PLL) modified QCM-D sensor, and the inhibition effect of resveratrol on FR expression levels on MCF-7 cells was monitored by chitosan-folic acid (CS-FA) composite membrane functionalized Au substrate for the first time. Changes in morphology and the cellular state of MCF-7 cells stimulated by resveratrol at different concentrations were detected by inverted fluorescence microscopy and flow cytometry. Atomic force microscopy confirmed that resveratrol influenced the cellular mechanical properties. The results indicated that the MCF-7 cells lose their original elasticity and increase their stiffness induced by resveratrol. It was further observed by confocal fluorescence imaging that resveratrol reduced the FR expression levels on the living cells surface. This study established a typical model of the QCM-D biosensor to evaluate the protein biomarker expression levels on the cells surface. QCM-D, which was used to investigate potential targets for an antitumor drug on living cells and realize a better understanding of the drug action mechanism, was expected to be developed into a promising tool for the screening of drugs.


Asunto(s)
Técnicas Biosensibles , Ensayos de Selección de Medicamentos Antitumorales , Receptores de Folato Anclados a GPI/biosíntesis , Modelos Biológicos , Tecnicas de Microbalanza del Cristal de Cuarzo , Estilbenos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Receptores de Folato Anclados a GPI/antagonistas & inhibidores , Humanos , Células MCF-7 , Microscopía Fluorescente , Resveratrol , Relación Estructura-Actividad
17.
Analyst ; 140(21): 7407-16, 2015 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-26405719

RESUMEN

Type 1 diabetes is an insulin-dependent metabolic disorder always associated with ketoacidosis and a high morbidity rate in teenagers. The in situ single molecule detection of insulin receptors on healthy and diseased erythrocytes is helpful to understand the pathomechanism of type 1 diabetes ketoacidosis (T1-DKA), which would also benefit the diagnosis and treatment of T1-DKA. Here, we demonstrated, for the first time, the single molecule interaction between insulin and insulin receptor on erythrocytes from a healthy volunteer and a T1-DKA patient using high sensitivity atomic force microscopy (AFM) in PBS solution. The single molecule force results demonstrated the decreased binding force and binding probability between insulin and insulin receptor on T1-DKA erythrocytes, implying the deficit of insulin receptor functions in T1-DKA. The binding kinetic parameters calculated from dynamic force spectroscopy indicated that the insulin-insulin receptor complexes on T1-DKA erythrocytes were less stable than those from healthy volunteer. Using high resolution AFM imaging, a decreased roughness was found both in intact T1-DKA erythrocytes and in the purified membrane of T1-DKA erythrocytes, and an increased stiffness was also found in T1-DKA erythrocytes. Moreover, AFM, which was used to investigate the single molecule interactions between insulin-insulin receptor, cell surface ultrastructure and stiffness in healthy and diseased erythrocytes, was expected to develop into a potential nanotool for pathomechanism studies of clinical samples at the nanoscale.


Asunto(s)
Antígenos CD/metabolismo , Diabetes Mellitus Tipo 1/sangre , Cetoacidosis Diabética/sangre , Eritrocitos/metabolismo , Microscopía de Fuerza Atómica/métodos , Receptor de Insulina/metabolismo , Adolescente , Separación Celular , Membrana Eritrocítica/metabolismo , Eritrocitos/citología , Humanos , Insulina/análisis , Insulina/química , Insulina/metabolismo , Masculino , Estrés Mecánico , Adulto Joven
18.
Bioorg Med Chem Lett ; 24(2): 679-84, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24365157

RESUMEN

Tumor angiogenesis is a complicated process based upon a sequence of interactions between tumor and vessel endothelial cells. Tumor conditioned medium has been widely used to stimulate endothelial cells in vitro angiogenesis. This work was aimed to investigate the effects of gold nanoparticles (GNPs) on angiogenesis in hepatic carcinoma-conditioned endothelial cells. Human umbilical vein endothelial cells (HUVECs) were cultured with conditioned medium (CM) from the human hepatocarcinoma cell line HepG2 (HepG2-CM), and then treated with different concentrations of GNPs. The effects of GNPs on the viability, migration and active VEGF level of HUVECs were investigated by MTT assay, wound healing assay and transwell chamber assay, and ELISA assay, respectively. The data showed that GNPs significantly inhibited HUVECs proliferation and migration induced by HepG2-CM, and also reduced the levels of active VEGF in the co-culture system. Then, the alterations in morphology and ultrastructure of HUVECs detected by atomic force microscopy (AFM) showed that there appeared obvious pseudopodia, larger membrane particle sizes and much rougher surface in HUVECs after HepG2-CM treatment, which were all reversed after GNPs treatment. Changes in cytoskeleton of HUVECs determined by immunocytochemistry demonstrated that GNPs treatment remarkably inhibited the activation effect of HepG2-CM on HUVECs, which was associated with the disruption of actin filaments induced by GNPs. This study indicates that GNPs can significantly inhibit HepG2-CM activated endothelial cell proliferation and migration through down-regulation of VEGF activity and disruption of cell morphology, revealing the potential applications of GNPs as antiangiogenic agent for the treatment of hepatic carcinoma.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Oro/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neoplasias Hepáticas Experimentales , Nanopartículas del Metal , Animales , Movimiento Celular/fisiología , Técnicas de Cocultivo/métodos , Oro/uso terapéutico , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neoplasias Hepáticas Experimentales/patología
19.
J Inflamm Res ; 17: 1397-1411, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476473

RESUMEN

Purpose: To investigate the correlation between M1/M2 macrophages (M1/M2 Mφ) and cell death mode under Mycobacterium tuberculosis (Mtb) infection. Methods: Raw gene expression profiles were collected from the Gene Expression Omnibus (GEO) database. Genes related to different cell death modes were collected from the KEGG, FerrDb and GSEA databases. The differentially expressed genes (DEGs) of the gene expression profiles were identified using the limma package in R. The intersection genes of M1/M2 Mφ with different cell death modes were obtained by the VennDiagram package. Hub genes were obtained by constructing the protein-protein interactions (PPI) network and Receiver Operating Characteristic (ROC) curve analysis. The expression of cell death modes marker genes and Hub genes were verified by Western Blot and Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). Results: Bioinformatics analysis was performed to screen Hub genes of Mtb-infected M1 Mφ and different cell death modes, naming NFKB1, TNF, CFLAR, TBK1, IL6, RELA, SOCS1, AIM2; Hub genes of Mtb-infected M2 Mφ and different cell death modes, naming TNF, BIRC3, MAP1LC3C, DEPTOR, UVRAG, SOCS1. Combined with experimental validation, M1 Mφ under Mtb infection showed higher expression of death (including apoptosis, autophagy, ferroptosis, and pyroptosis) genes compared to M2 Mφ and genes such as NFKB1, TNF, CFLAR, TBK1, IL6, RELA, AIM2, BIRC3, DEPTOR show differential expression. Conclusion: NFKB1, TNF, CFLAR, TBK1, IL6, RELA, AIM2 in Mtb-infected M1 Mφ, and TNF, BIRC3, DEPTOR in Mtb-infected M2 Mφ might be used as potential diagnostic targets for TB. At early stage of Mtb infection, apoptosis, autophagy, ferroptosis, and pyroptosis occurred more significantly in M1 Mφ than that in M2 Mφ, which may contribute to the transition of Mtb-infected Mφ from M1-dominant to M2-dominant and contribute to the immune escape mechanisms of Mtb.

20.
Curr Med Chem ; 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38231073

RESUMEN

Iron, copper, and zinc play integral roles in the battle against Mycobacterium tuberculosis (Mtb) infection; however, they are often trapped between nutrients and toxins, posing a significant challenge to macrophages and Mtb to utilize them. Due to this two-sided effect, macrophages and Mtb strictly regulate metal uptake, storage, and excretion. This review discusses the balanced regulation of iron, copper, and zinc in macrophages and Mtb during infection, focusing on the intracellular metal regulatory system. Macrophages typically use the two-sided effect of metals to limit Mtb access to nutrients or poison them. Mtb has developed a metal metabolism regulatory mechanism compatible with the nutritional immune strategy. This includes the mediation of relevant metalloproteins and metalloenzymes to maintain the multimetal balance. This review also explored the regulation of metal metabolism homeostasis in macrophages resistant to Mtb infection, providing a theoretical foundation for identifying potential clinical targets for Mtb infection, developing metalloid anti-tuberculosis drugs, and understanding the immune mechanisms against intracellular Mtb infection.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA