Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Autoimmun ; 100: 84-94, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30872080

RESUMEN

TREX1 encodes a major cellular DNA exonuclease. Mutations of this gene in human cause cellular accumulation of DNA that triggers autoimmune diseases including Aicardi-Goutieres Syndrome (AGS) and systemic lupus erythematosus (SLE). We created a lupus mouse model by engineering a D18 N mutation in the Trex1 gene which inactivates the enzyme and has been found in human patients with lupus-like disorders. The Trex1D18N/D18N mice exhibited systemic inflammation that consistently recapitulates many characteristics of human AGS and SLE. Importantly, ablation of cGas gene in the Trex1D18N/D18N mice rescued the lethality and all detectable pathological phenotypes, including multi-organ inflammation, interferon stimulated gene induction, autoantibody production and aberrant T-cell activation. These results indicate that cGAS is a key mediator in the autoimmune disease associated with defective TREX1 function, providing additional insights into disease pathogenesis and guidance to the development of therapeutics for human systemic autoimmune disorders.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Exodesoxirribonucleasas , Lupus Eritematoso Sistémico , Activación de Linfocitos , Mutación Missense , Malformaciones del Sistema Nervioso , Nucleotidiltransferasas , Fosfoproteínas , Linfocitos T/inmunología , Sustitución de Aminoácidos , Animales , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/patología , Modelos Animales de Enfermedad , Activación Enzimática/genética , Activación Enzimática/inmunología , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/inmunología , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Mutantes , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/inmunología , Malformaciones del Sistema Nervioso/patología , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Linfocitos T/patología
2.
Wei Sheng Wu Xue Bao ; 56(1): 19-25, 2016 Jan 04.
Artículo en Zh | MEDLINE | ID: mdl-27305776

RESUMEN

Epstein-Barr virus (EBV) is the first identified human oncogenic DNA virus in the gamma-herpesvirus family. EBV triggers a cascade events of innate immune responses through Toll-like receptor signaling including the production of type I interferons and the activation of functional autophagy. However, EBV has developed much more elaborate and sophisticated strategies for subverting and escaping the host immune system, such as limiting its own gene expression, activing the host ubiquitin-specific protease system, and interfering ubiquitin modification. EBV impairs the host immune system, leading to lifelong persistent infections, which in turn result in the occurrence of EBV-associated diseases, such as nasopharyngeal carcinoma and infectious mononucleosis. Thus, to better understand the mechanisms regarding the infection latentency and oncogenicity of EBV invasion will be crucial for identifying potential immunotherapeutic targets for EBV- related diseases, such as infectious mononucleosis and nasopharyngeal carcinoma. In this article, we discuss the research advances regarding the virology and immunology of EBV in the modulation of the host immune response and evasion.


Asunto(s)
Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Animales , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata
3.
Biomed Pharmacother ; 157: 114004, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36375308

RESUMEN

When the production of reactive oxygen species (ROS) is overloaded surpassing the capacity of the reductive rheostat, mammalian cells undergo a series of oxidative damage termed oxidative stress (OS). This phenomenon is ubiquitously detected in many human pathological conditions. Wound healing program implicates continuous neovascularization, cell proliferation, and wound remodeling. Increasing evidence indicates that reactive oxygen species (ROS) have profound impacts on the wound healing process through regulating a series of the physiological and pathological program including inflammatory response, cell proliferation, angiogenesis, granulation as well as extracellular matrix formation. In most pathological wound healing processes, excessive ROS exerts a negative role on the wound healing process. Interestingly, the moderate increase of ROS levels is beneficial in killing bacteria at the wound site, which creates a sterile niche for revascularization. In this review, we discussed the physiological rhythms of wound healing and the role of ROS in this progress, aim to explore the potential manipulation of OS as a promising therapeutic avenue.


Asunto(s)
Estrés Oxidativo , Cicatrización de Heridas , Animales , Humanos , Especies Reactivas de Oxígeno , Cicatrización de Heridas/fisiología , Proliferación Celular , Neovascularización Patológica , Mamíferos
4.
iScience ; 26(7): 107090, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37416470

RESUMEN

TREX1 encodes a major DNA exonuclease and mutations of this gene are associated with type I interferonopathies in human. Mice with Trex1 deletion or mutation have shortened life spans accompanied by a senescence-associated secretory phenotype. However, the contribution of cellular senescence in TREX1 deficiency-induced type I interferonopathies remains unknown. We found that features of cellular senescence present in Trex1-/- mice are induced by multiple factors, particularly DNA damage. The cGAS-STING and DNA damage response pathways are required for maintaining TREX1 deletion-induced cellular senescence. Inhibition of the DNA damage response, such as with Checkpoint kinase 2 (CHK2) inhibitor, partially alleviated progression of type I interferonopathies and lupus-like features in the mice. These data provide insights into the initiation and development of type I interferonopathies and lupus-like diseases, and may help inform the development of targeted therapeutics.

5.
ACS Appl Mater Interfaces ; 14(37): 42541-42557, 2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36094305

RESUMEN

Nanotechnology has shown a revolution in cancer treatments, including breast cancers. However, there remain some challenges and translational hurdles. Surgery, radiotherapy, and chemotherapy are the primary treatment methods for breast cancer, although drug combinations showed promising results in preclinical studies. Herein we report the development of a smart drug delivery system (DDS) to efficiently treat breast cancer by pyroptosis-starvation-chemotherapeutic combination. Cancer-starvation agent glucose oxidase was chemically attached to synthesized iron oxide nanoparticles which were entrapped inside poly(lactic-co-glycolic acid) along with apoptosis-associated speck-like protein containing a caspase recruitment domain plasmid and paclitaxel (PTX). An emulsion solvent evaporation method was used to prepare the DDS. The surface of the DDS was modified with chitosan to which aptamer was attached to achieve site-specific targeting. Hence, the prepared DDS could be targeted to a tumor site by both external magnet and aptamer to obtain an enhanced accumulation of drugs at the tumor site. The final size of the aptamer-decorated DDS was less than 200 nm, and the encapsulation efficiency of PTX was 76.5 ± 2.5%. Drug release from the developed DDS was much higher at pH 5.5 than at pH 7.4, ensuring the pH sensitivity of the DDS. Due to efficient dual targeting of the DDS, in vitro viability of 4T1 cells was reduced to 12.1 ± 1.6%, whereas the nontargeted group and free PTX group could reduce the viability of cells to 29.2 ± 2.4 and 46.2 ± 1.6%, respectively. Our DDS showed a synergistic effect in vitro and no severe side effects in vivo. This DDS has strong potential to treat various cancers.


Asunto(s)
Neoplasias de la Mama , Quitosano , Nanopartículas , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quitosano/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Emulsiones , Femenino , Glucosa Oxidasa/uso terapéutico , Humanos , Fenómenos Magnéticos , Nanopartículas/química , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico , Polímeros/química , Piroptosis , Solventes
6.
Biochem Pharmacol ; 178: 114090, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32565148

RESUMEN

Celastrol is one of most potent bioactive molecule isolated from the medicinal plant Tripterygium wilfordii (Thunder God Vine) and is well known for its potential therapeutic value against various chronic diseases including the autoimmune diseases, such as systemic lupus erythematosus and Aicardi-Goutieres syndrome, or other interferonopathies. However, the underlying mechanism of celastrol function remains unclear. Here we showed that celastrol caused inhibition of interferon regulatory factor 3 (IRF3) activation leading to the down-regulation of the interferon response triggered by cytosolic nucleic acids in vitro and in vivo. Moreover, celastrol treatment markedly ameliorates the autoimmune phenotypes including myocarditis, aberrant interferon response and autoantibody production, as well as the excessive T-cell activation in Trex1-/- autoimmune disease mouse model. Collectively, our results indicate that celastrol inhibits interferon response by targeting IRF3 activation and may be used as an effective treatment for interferon response-dependent autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/metabolismo , Exodesoxirribonucleasas/deficiencia , Fosfoproteínas/deficiencia , Tripterygium , Triterpenos/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Triterpenos Pentacíclicos , Células RAW 264.7 , Distribución Aleatoria , Triterpenos/aislamiento & purificación
7.
J Cancer ; 10(27): 6925-6932, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31839828

RESUMEN

Purpose: Recent studies indicate that pregnancy upregulated non-ubiquitous calmodulin kinase (PNCK) is significantly up-regulated in breast and renal carcinomas. However, the expression profile and its biological relevance of PNCK in nasopharyngeal carcinoma (NPC) have not been elucidated. Methods: The expression level of PNCK was detected in specimens of NPC (n=10) and normal tissues (n=10) by real-time PCR and immunohistochemistry. Celigo Cell Counting and MTT assay were used to measure cell viability. Apoptosis was detected by flow cytometric analysis and caspases 3/7 activity assay. Real-time PCR and Western blotting were performed to evaluate the expression of PNCK. The bioluminescence imaging was used to evaluate the effects of PNCK knockdown on tumor growth using a xenograft animal model. The global gene expression profile was determined in wild type and PNCK-depleted CNE-2 cells via transcriptomics analysis. For mechanical investigation, the changes of PI3K/AKT/mTOR signaling pathway were detected by Western blotting. Results: The mRNA and protein levels of PNCK were increased in human NPC samples. In vitro experiments showed that shRNA or CRISPR-Cas9 mediated silencing of PNCK inhibited proliferation and induced apoptosis in NPC cells. In addition, in vivo assay revealed that knockdown of PNCK suppressed tumor growth. Consistently, a significant reduction of tumor bioluminescence in mice inoculated with PNCK-knockdown cells compared to that of control cells. In gene expression, the transcriptomics analysis revealed that there were 589 upregulated genes and 589 downregulated genes in PNCK-knockdown cells. Ingenuity Pathway Analysis (IPA) identified significant changes of PI3K/AKT/mTOR signaling pathway in PNCK-knockdown cells. Furthermore, western blot analysis revealed that interference with PNCK reduced the phosphorylation levels of PI3K, AKT and mTOR in CNE-2 cells. Conclusion: This study for the first time demonstrates that knockdown of PNCK could suppress growth and induce apoptosis of NPC cells both in vitro and in vivo by regulating PI3K/AKT/mTOR signaling pathway. These findings suggest that PNCK might be a novel therapeutic target for NPC treatment.

8.
J Biomed Opt ; 21(12): 125003, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27936269

RESUMEN

Surface-enhanced Raman spectroscopy (SERS) was employed to detect deoxyribose nucleic acid (DNA) variations associated with the development of nasopharyngeal carcinoma (NPC). Significant SERS spectral differences between the DNA extracted from early NPC, advanced NPC, and normal nasopharyngeal tissue specimens were observed at 678, 729, 788, 1337, 1421, 1506, and 1573??cm?1, which reflects the genetic variations in NPC. Principal component analysis combined with discriminant function analysis for early NPC discrimination yielded a diagnostic accuracy of 86.8%, 92.3%, and 87.9% for early NPC, advanced NPC, and normal nasopharyngeal tissue DNA, respectively. In this exploratory study, we demonstrated the potential of SERS for early detection of NPC based on the DNA molecular study of biopsy tissues.


Asunto(s)
Carcinoma/diagnóstico por imagen , ADN/análisis , Interpretación de Imagen Asistida por Computador/métodos , Neoplasias Nasofaríngeas/diagnóstico por imagen , Espectrometría Raman/métodos , Algoritmos , Carcinoma/química , ADN/química , Humanos , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/química , Análisis de Componente Principal , Plata/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA