Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 590(7846): 410-415, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33597760

RESUMEN

Current X-ray imaging technologies involving flat-panel detectors have difficulty in imaging three-dimensional objects because fabrication of large-area, flexible, silicon-based photodetectors on highly curved surfaces remains a challenge1-3. Here we demonstrate ultralong-lived X-ray trapping for flat-panel-free, high-resolution, three-dimensional imaging using a series of solution-processable, lanthanide-doped nanoscintillators. Corroborated by quantum mechanical simulations of defect formation and electronic structures, our experimental characterizations reveal that slow hopping of trapped electrons due to radiation-triggered anionic migration in host lattices can induce more than 30 days of persistent radioluminescence. We further demonstrate X-ray luminescence extension imaging with resolution greater than 20 line pairs per millimetre and optical memory longer than 15 days. These findings provide insight into mechanisms underlying X-ray energy conversion through enduring electron trapping and offer a paradigm to motivate future research in wearable X-ray detectors for patient-centred radiography and mammography, imaging-guided therapeutics, high-energy physics and deep learning in radiology.

2.
J Biol Chem ; 299(5): 104613, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36931391

RESUMEN

Epstein-Barr virus (EBV) is a member of the lymphotropic virus family and is highly correlated with some human malignant tumors. It has been reported that envelope glycoprotein 110 (gp110) plays an essential role in viral fusion, DNA replication, and nucleocapsid assembly of EBV. However, it has not been established whether gp110 is involved in regulating the host's innate immunity. In this study, we found that gp110 inhibits tumor necrosis factor α-mediated NF- κB promoter activity and the downstream production of NF- κB-regulated cytokines under physiological conditions. Using dual-luciferase reporter assays, we showed that gp110 might impede the NF-κB promoter activation downstream of NF-κB transactivational subunit p65. Subsequently, we used coimmunoprecipitation assays to demonstrate that gp110 interacts with p65 during EBV lytic infection, and that the C-terminal cytoplasmic region of gp110 is the key interaction domain with p65. Furthermore, we determined that gp110 can bind to the N-terminal Rel homologous and C-terminal domains of p65. Alternatively, gp110 might not disturb the association of p65 with nontransactivational subunit p50, but we showed it restrains activational phosphorylation (at Ser536) and nuclear translocation of p65, which we also found to be executed by the C-terminal cytoplasmic region of gp110. Altogether, these data suggest that the surface protein gp110 may be a vital component for EBV to antagonize the host's innate immune response, which is also helpful for revealing the infectivity and pathogenesis of EBV.


Asunto(s)
Infecciones por Virus de Epstein-Barr , FN-kappa B , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Herpesvirus Humano 4/metabolismo , Infecciones por Virus de Epstein-Barr/genética , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transducción de Señal , Transporte de Proteínas
3.
Inflamm Res ; 73(1): 47-63, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38147126

RESUMEN

OBJECTIVE: NLRP3 inflammasome-mediated pyroptosis of macrophage acts essential roles in the progression of sepsis-induced acute lung injury (ALI). Tangeretin (TAN), enriched in citrus fruit peel, presents anti-oxidative and anti-inflammatory effects. Here, we aimed to explore the potentially protective effect of TAN on sepsis-induced ALI, and the underlying mechanism of TAN in regulating NLRP3 inflammasome. MATERIAL AND METHODS: The effect of TAN on sepsis-induced ALI and NLRP3 inflammasome-mediated pyroptosis of macrophage were examined in vivo and in vitro using a LPS-treated mice model and LPS-induced murine macrophages, respectively. The mechanism of TAN regulating the activation of NLRP3 inflammasome in sepsis-induced ALI was investigated with HE staining, Masson staining, immunofluorescent staining, ELISA, molecular docking, transmission electron microscope detection, qRT-PCR, and western blot. RESULTS: TAN could evidently attenuate sepsis-induced ALI in mice, evidenced by reducing pulmonary edema, pulmonary congestion and lung interstitial fibrosis, and inhibiting macrophage infiltration in the lung tissue. Besides, TAN significantly suppressed inflammatory cytokine IL-1ß and IL-18 expression in the serum or bronchoalveolar lavage fluid (BALF) samples of mice with LPS-induced ALI, and inhibited NLRP3 inflammasome-mediated pyroptosis of macrophages. Furthermore, we found TAN inhibited ROS production, preserved mitochondrial morphology, and alleviated excessive mitochondrial fission in LPS-induced ALI in mice. Through bioinformatic analysis and molecular docking, Polo-like kinase 1 (PLK1) was identified as a potential target of TAN for treating sepsis-induced ALI. Moreover, TAN significantly inhibited the reduction of PLK1 expression, AMP-activated protein kinase (AMPK) phosphorylation, and Dynamin related protein 1 (Drp1) phosphorylation (S637) in LPS-induced ALI in mice. In addition, Volasertib, a specific inhibitor of PLK1, abolished the protective effects of TAN against NLRP3 inflammasome-mediated pyroptosis of macrophage and lung injury in the cell and mice septic models. CONCLUSION: TAN attenuates sepsis-induced ALI by inhibiting ROS-mediated NLRP3 inflammasome activation via regulating PLK1/AMPK/DRP1 signaling axis, and TAN is a potentially therapeutic candidate against ALI through inhibiting pyroptosis.


Asunto(s)
Lesión Pulmonar Aguda , Sepsis , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas Activadas por AMP , Lipopolisacáridos/farmacología , Simulación del Acoplamiento Molecular , Lesión Pulmonar Aguda/inducido químicamente , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Ratones Endogámicos C57BL
4.
Mikrochim Acta ; 191(6): 311, 2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38717575

RESUMEN

Urine retinol-binding protein 4 (RBP4) has recently been reported as a novel earlier biomarker of chronic kidney disease (CKD) which is a global public health problem with high morbidity and mortality. Accurate and rapid detection of urine RBP4 is essential for early monitor of impaired kidney function and prevention of CKD progression. In the present study, we developed a time-resolved fluorescence immunochromatographic test strip (TRFIS) for the quantitative and rapid detection of urine RBP4. This TRFIS possessed excellent linearity ranging from 0.024 to 12.50 ng/mL for the detection of urine RBP4, and displayed a good linearity (Y = 239,581 × X + 617,238, R2 = 0.9902), with the lowest visual detection limit of 0.049 ng/mL. This TRFIS allows for quantitative detection of urine RBP4 within 15 min and shows high specificity. The intra-batch coefficient of variation (CV) and the inter-batch CV were both < 8%, respectively. Additionally, this TRFIS was applied to detect RBP4 in the urine samples from healthy donors and patients with CKD, and the results of TRFIS could efficiently discern the patients with CKD from the healthy donors. The developed TRFIS has the characteristics of high sensitivity, high accuracy, and a wide linear range, and is suitable for rapid and quantitative determination of urine RBP4.


Asunto(s)
Cromatografía de Afinidad , Insuficiencia Renal Crónica , Proteínas Plasmáticas de Unión al Retinol , Humanos , Proteínas Plasmáticas de Unión al Retinol/orina , Cromatografía de Afinidad/métodos , Insuficiencia Renal Crónica/orina , Insuficiencia Renal Crónica/diagnóstico , Límite de Detección , Tiras Reactivas , Biomarcadores/orina , Inmunoensayo/métodos
5.
Mikrochim Acta ; 191(6): 325, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38739279

RESUMEN

Glial fibrillary acidic protein (GFAP) in serum has been shown as a biomarker of traumatic brain injury (TBI) which is a significant global public health concern. Accurate and rapid detection of serum GFAP is critical for TBI diagnosis. In this study, a time-resolved fluorescence immunochromatographic test strip (TRFIS) was proposed for the quantitative detection of serum GFAP. This TRFIS possessed excellent linearity ranging from 0.05 to 2.5 ng/mL for the detection of serum GFAP and displayed good linearity (Y = 598723X + 797198, R2 = 0.99), with the lowest detection limit of 16 pg/mL. This TRFIS allowed for quantitative detection of serum GFAP within 15 min and showed high specificity. The intra-batch coefficient of variation (CV) and the inter-batch CV were both < 4.0%. Additionally, this TRFIS was applied to detect GFAP in the serum samples from healthy donors and patients with cerebral hemorrhage, and the results of TRFIS could efficiently discern the patients with cerebral hemorrhage from the healthy donors. Our developed TRFIS has the characteristics of high sensitivity, high accuracy, and a wide linear range and is suitable for rapid and quantitative determination of serum GFAP on-site.


Asunto(s)
Cromatografía de Afinidad , Proteína Ácida Fibrilar de la Glía , Humanos , Biomarcadores/sangre , Hemorragia Cerebral/sangre , Hemorragia Cerebral/diagnóstico , Cromatografía de Afinidad/métodos , Proteína Ácida Fibrilar de la Glía/sangre , Límite de Detección , Tiras Reactivas
6.
J Transl Med ; 21(1): 652, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37737187

RESUMEN

BACKGROUND: Parkinson's disease (PD), characterized by the progressive loss of dopaminergic neurons in the substantia nigra and striatum of brain, seriously threatens human health, and is still lack of effective treatment. Dysregulation of N6-methyladenosine (m6A) modification has been implicated in PD pathogenesis. However, how m6A modification regulates dopaminergic neuronal death in PD remains elusive. Mesenchymal stem cell-derived exosomes (MSC-Exo) have been shown to be effective for treating central nervous disorders. We thus propose that the m6A demethylase FTO-targeted siRNAs (si-FTO) may be encapsulated in MSC-Exo (Exo-siFTO) as a synergistic therapy against dopaminergic neuronal death in PD. METHODS: In this study, the effect of m6A demethylase FTO on dopaminergic neuronal death was evaluated both in vivo and in vitro using a MPTP-treated mice model and a MPP + -induced MN9D cellular model, respectively. The mechanism through which FTO influences dopaminergic neuronal death in PD was investigated with qRT-PCR, western blot, immumohistochemical staining, immunofluorescent staining and flow cytometry. The therapeutic roles of MSC-Exo containing si-FTO were examined in PD models in vivo and in vitro. RESULTS: The total m6A level was significantly decreased and FTO expression was increased in PD models in vivo and in vitro. FTO was found to promote the expression of cellular death-related factor ataxia telangiectasia mutated (ATM) via m6A-dependent stabilization of ATM mRNA in dopaminergic neurons. Knockdown of FTO by si-FTO concomitantly suppressed upregulation of α-Synuclein (α-Syn) and downregulation of tyrosine hydroxylase (TH), and alleviated neuronal death in PD models. Moreover, MSC-Exo were utilized to successfully deliver si-FTO to the striatum of animal brain, resulting in the significant suppression of α-Syn expression and dopaminergic neuronal death, and recovery of TH expression in the brain of PD mice. CONCLUSIONS: MSC-Exo delivery of si-FTO synergistically alleviates dopaminergic neuronal death in PD via m6A-dependent regulation of ATM mRNA.


Asunto(s)
Ataxia Telangiectasia , Exosomas , Enfermedad de Parkinson , Humanos , Animales , Ratones , ARN Interferente Pequeño , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia , ARN Mensajero/genética , Neuronas Dopaminérgicas , Dopamina , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Proteínas de la Ataxia Telangiectasia Mutada
7.
J Integr Neurosci ; 22(6): 138, 2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-38176925

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a common progressive neurodegenerative disease. The Ubiquitin-Protease system (UPS), which plays important roles in maintaining protein homeostasis in eukaryotic cells, is involved in the development of AD. This study sought to identify differential UPS-related genes (UPGs) in AD patients by using bioinformatic methods, reveal potential biomarkers for early detection of AD, and investigate the association between the identified biomarkers and immune cell infiltration in AD. METHODS: The differentially expressed UPGs were screened with bioinformatics analyses using the Gene Expression Omnibus (GEO) database. A weighted gene co-expression network analysis (WGCNA) analysis was performed to explore the key gene modules associated with AD. A Single-sample Gene Set Enrichment Analysis (ssGSEA) analysis was peformed to explore the patterns of immune cells in the brain tissue of AD patients. Real-time quantitative PCR (RT-qPCR) was performed to examine the expression of hub genes in blood samples from healthy controls and AD patients. RESULTS: In this study, we identified four UPGs (USP3, HECW2, PSMB7, and UBE2V1) using multiple bioinformatic analyses. Furthermore, three UPGs (USP3, HECW2, PSMB7) that are strongly correlated with the clinical features of AD were used to construct risk score prediction markers to diagnose and predict the severity of AD. Subsequently, we analyzed the patterns of immune cells in the brain tissue of AD patients and the associations between immune cells and the three key UPGs. Finally, the risk score model was verified in several datasets of AD and showed good accuracy. CONCLUSIONS: Three key UPGs are identified as potential biomarker for AD patients. These genes may provide new targets for the early identification of AD patients.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Humanos , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Biomarcadores , Biología Computacional , Ubiquitinas , Ubiquitina-Proteína Ligasas , Proteasas Ubiquitina-Específicas
8.
Clin Immunol ; 245: 109176, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36368640

RESUMEN

Atherosclerosis, characterized as the chronic inflammation of the arterial wall, is one of the leading causes of coronary artery disease (CAD), and macrophages are found to play essential roles in the initiation and progression of inflammation in atherosclerosis. N6-methyladenosine (m6A) modification, as the most abundant epi-transcriptomic modification in mRNA, is found to mediate the atherogenic inflammatory cascades in vascular endothelium. The detailed molecular mechanism of m6A methylation regulating inflammatory response during atherosclerosis is still not fully known. In this study, we find oxidized low-density lipoprotein (oxLDL) stimulation increases methyltransferases Mettl3 and Mettl14 expressions in macrophages, whereas the total m6A modification level in macrophages decreases under oxLDL stimulation. Matrin-3 (Matr3), an RNA binding protein, is identified to play a suppressive role on oxLDL-mediated macrophage inflammatory responses through inhibiting activation of pro-inflammatory signaling, mitogen-activated protein kinase (Mapk) by m6A-mediated mRNA decay via regulating the formation of Mettl3-Mettl14 complex. Moreover, we find that Matr3 expression decreases in the oxLDL-stimulated macrophages, and the peripheral blood-derived monocytes from patients with CAD, and overexpression of Matr3 significantly alleviates atherosclerosis development in vivo. Our study for the first time clarifies the role of Matr3 on macrophage inflammatory responses during atherosclerotic development, and supplies deep understanding on the relationship of m6A modification and inflammatory responses in atherosclerosis.


Asunto(s)
Aterosclerosis , Metiltransferasas , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas de Unión al ARN , Inflamación , Macrófagos/metabolismo , Aterosclerosis/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo
9.
Exp Cell Res ; 396(2): 112332, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33065113

RESUMEN

DEAD-box (DDX) helicases are critical for recognizing viral nucleic acids to regulate antiviral innate immunity. Although DDX5 has been reported to participate in various virus infection, whether DDX5 regulates innate immune responses and its underlying mechanisms are still unknown. Here, we report that DDX5 is a negative regulator of type I IFN (IFN-I) production in antiviral responses. DDX5 knockdown significantly promoted DNA or RNA virus infection-induced IFN-I production and IFN-stimulated genes (ISGs) expression, while ectopic expression of DDX5 inhibited IFN-I production and promoted viral replication. Furthermore, we found that DDX5 specifically interacted with serine/threonine-protein phosphatase 2 A catalytic subunit beta (PP2A-Cß) and viral infection enhanced the interaction between DDX5 and PP2A-Cß. Besides, PP2A-Cß interacted with IFN regulatory factor 3 (IRF3), and PP2A-Cß knockdown promoted viral infection-induced IRF3 phosphorylation and IFN-I production. In addition, DDX5 knockdown rendered the mice more resistant to viral infection and enhanced antiviral innate immunity in vivo. Thus, DDX5 suppresses IFN-I antiviral innate immune response by interacting with PP2A-Cß to deactivate IRF3. Together, these findings identify a negative role of DDX5 on regulating IFN-I signaling in innate immune responses.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Proteína Fosfatasa 2/metabolismo , Animales , Femenino , Técnicas de Silenciamiento del Gen , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Regulación hacia Arriba/genética , Virosis/metabolismo , Virosis/patología
10.
J Stroke Cerebrovasc Dis ; 29(6): 104801, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32249206

RESUMEN

BACKGROUND: Ischemic stroke is the leading cause of disability and death globally. Micro-RNAs (miRNAs) have been reported to play important roles in the development and pathogenesis of the nervous system. However, the exact function and mechanism of miRNAs have not been fully elucidated about brain damage caused by cerebral ischemia/reperfusion (I/R). METHODS: In this study, we explored the neuroprotective effects of miR-219a-5p on brain using an in vitro ischemia model (mouse neuroblastoma N2a cells treated with oxyglucose deprivation and reperfusion), and in vivo cerebral I/R model in mice. Western blot assay and Reverse Transcription-Polymerase Chain Reaction were used to check the expression of molecules involved. Flow cytometry and cholecystokinin were used to examine cell apoptosis, respectively. RESULTS: Our research shows that miR-219a-5p gradually decreases in cerebral I/R models in vivo and in vitro. In vitro I/R, we find that miR-219a-5p mimics provided evidently protection for cerebral I/R damage, as shown by increased cell viability and decreased the release of LDH and cell apoptosis. Mechanically, our findings indicate that miR-219a-5p binds to cAMP specific 3', 5'-cyclic phosphodiesterase 4D (PDE4D) mRNA in the 3'-UTR region, which subsequently leads to a decrease in Pde4d expression in I/R N2a cells. CONCLUSIONS: Our results provide new ideas for the study of the mechanism of cerebral ischemia/reperfusion injury, and lay the foundation for further research on the treatment of brain I/R injury. Upregulation of miR-219a-5p decreases cerebral ischemia/reperfusion injury by targeting Pde4d in vitro.


Asunto(s)
Apoptosis , Encéfalo/enzimología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Infarto de la Arteria Cerebral Media/enzimología , MicroARNs/metabolismo , Neuronas/enzimología , Daño por Reperfusión/enzimología , Regiones no Traducidas 3' , Animales , Sitios de Unión , Encéfalo/patología , Línea Celular Tumoral , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Neuronas/patología , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Transducción de Señal
11.
Anal Chem ; 91(15): 10149-10155, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31305067

RESUMEN

Optical biosensors that enable highly sensitive detection of biomolecules are useful for applications in early disease diagnosis. However, the presence of UV-vis-induced background fluorescence in biological samples is still challenging. Thanks to the weak scattering and nearly no absorption of biological chromophores under X-ray excitation, we describe the development of an X-ray nanocrystal scintillator-based aptasensor that is able to achieve sensitive and homogeneous detection of target biomolecules. In this work, aptamer-labeled lanthanide-doped nanocrystal scintillators was designed to rapidly and sensitively detect lysozyme via fluorescence resonance energy transfer (FRET) in human serum samples. Benefiting from the use of low-dose X-ray as an excitation source and high-efficiency luminescence of heavy atoms-contained nanocrystals, the proposed X-ray nanocrystal scintillator-based aptasensor can readily detect lysozyme with a high sensitivity up to 0.94 nM, as well as an excellent specificity and sample recoveries. Thus, our technique suggests that the X-ray scintillating aptasensor can create a new generation of autofluorescence-free high-sensitivity strategy for biomarker sensing in biomedical applications.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Fluorescencia , Muramidasa/sangre , Nanopartículas/química , Humanos , Rayos X
12.
J Stroke Cerebrovasc Dis ; 28(1): 175-184, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30539754

RESUMEN

BACKGROUND: Stevioside, isolated from the herb Stevia rebaudiana, has been widely used as a food sweetener all over the world. Isosteviol Sodium (STV-Na), an injectable formulation of isosteviol sodium salt, has been proved to possess much greater solubility and bioavailability and exhibit protective effects against cerebral ischemia injury in vivo by inhibiting neuron apoptosis. However, the underlying mechanisms of the neuroprotective effects STV-Na are still not completely known. In the present study, we investigated the effects of STV-Na on neuronal cell death caused by hypoxia in vitro and its underlying mechanisms. METHODS: We used cobalt chloride (CoCl2) to expose mouse neuroblastoma N2a cells to hypoxic conditions in vitro. RESULTS: Our results showed that pretreatment with STV-Na (20 µM) significantly attenuated the decrease of cell viability, lactate dehydrogenase release and cell apoptosis under conditions of CoCl2-induced hypoxia. Meanwhile, STV-Na pretreatment significantly attenuated the upregulation of intracellular Ca2+ concentration and reactive oxygen species production, and inhibited mitochondrial depolarization in N2a cells under conditions of CoCl2-induced hypoxia. Furthermore, STV-Na pretreatment significantly downregulated expressions of nitric oxide synthase, interleukin-1ß, tumor necrosis factor-α, interleukin-6, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinase (MAPK) signalings in N2a cells under conditions of CoCl2-induced hypoxia. CONCLUSIONS: Taken together, STV-Na protects neural cells against hypoxia-induced apoptosis through inhibiting MAPK and NF-κB pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Diterpenos de Tipo Kaurano/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/fisiología , Calcio/metabolismo , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cobalto/toxicidad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Neuronas/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Cell Physiol Biochem ; 44(2): 607-617, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29161701

RESUMEN

BACKGROUND/AIMS: Type I interferon (IFN-1) production and IFN-1 signaling play critical roles in the host antiviral innate immune responses. Although transcription factor Yin Yang 1 (YY1) has been reported to have a dual activator/repressor role during the regulation of interferon beta (IFN-ß) promoter activity, the roles of YY1 in the regulation of upstream signaling pathways leading to IFN-1 induction and IFN-1 signaling during viral infection remain to be elucidated. METHODS: The roles of YY1 in IFN-1 production and IFN-1 signaling were investigated using immunoblotting, real-time PCR, small interfering RNA (siRNA)-mediated YY1 knockdown, YY1 overexpression by transient transfection, and co-immunoprecipitation, using mouse cells. RESULTS: YY1 was shown to interact with STAT1 in the absence of viral infection. Following viral infection, YY1 protein expression levels were decreased. YY1 knockdown led to a considerable downregulation of phosphorylated (p) TBK1 and pIRF3 expressions, while YY1 overexpression significantly upregulated pTBK1 and pIRF3 expression levels and promoted virus-induced IFN-ß production. Additionally, YY1 knockdown led to a significant upregulation of pSTAT1, pSTAT2 and antiviral interferon-stimulated genes, and inhibited viral replication. CONCLUSION: We demonstrated here that YY1 interacts with STAT1 and dynamically regulates the induction of IFN-1 production and activation of IFN-1 signaling in different stages during viral infection.


Asunto(s)
Inmunidad Innata , Factor de Transcripción YY1/metabolismo , Animales , Línea Celular , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Inmunoprecipitación , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/análisis , Interferón beta/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Simplexvirus/fisiología , Transfección , Regulación hacia Arriba , Vesiculovirus/fisiología , Factor de Transcripción YY1/antagonistas & inhibidores , Factor de Transcripción YY1/genética
14.
Exp Cell Res ; 347(1): 83-94, 2016 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-27426727

RESUMEN

Apoptosis, as an innate antiviral defense, not only functions to limit viral replication by eliminating infected cells, but also contribute to viral dissemination, particularly at the late stages of infection. A highly neurotropic CVS strain of rabies virus induces apoptosis both in vitro and in vivo. However, the detailed mechanism of CVS-mediated neuronal apoptosis is not entirely clear. Here, we show that CVS induces apoptosis through mitochondrial pathway by dissipating mitochondrial membrane potential, release of cytochrome c and AIF. CVS blocks Bax activation at the early stages of infection; while M protein partially targets mitochondria and induces mitochondrial apoptosis at the late stages of infection. The α-helix structure spanning 67-79 amino acids of M protein is essential for mitochondrial targeting and induction of apoptosis. These results suggest that CVS functions on mitochondria to regulate apoptosis at different stages of infection, so as to for viral replication and dissemination.


Asunto(s)
Apoptosis , Mitocondrias/metabolismo , Virus de la Rabia/metabolismo , Proteínas de la Matriz Viral/metabolismo , Animales , Factor Inductor de la Apoptosis/metabolismo , Caspasa 9/metabolismo , Línea Celular , Activación Enzimática , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Rabia/metabolismo , Rabia/virología , Transducción de Señal , Proteína X Asociada a bcl-2/metabolismo
15.
J Stroke Cerebrovasc Dis ; 26(11): 2603-2614, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28784277

RESUMEN

BACKGROUND: Isosteviol sodium (STVNa) has been reported to have neuroprotective effects against ischemia/reperfusion (I/R) injury in rats. Furthermore, recanalization treatments, including thrombolytic therapy, have several limitations. Excessive inflammation and apoptosis contribute to the pathogenesis of ischemic brain damage. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is critical to these processes and is associated with cerebral ischemia. Therefore, we studied the potential therapeutic effects and mechanisms of STVNa on permanent cerebral ischemia in mice. METHODS: Permanent middle cerebral artery occlusion (pMCAO) was established via the suture method, followed by intravenous STVNa (7.5, 15, 30, 45, and 60 mg/kg). Neurobehavioral deficits, infarct volume, and histology were examined 24 hours after cerebral ischemia. In addition, the messenger RNA (mRNA) expression of NF-κB-related genes was detected using real-time quantitative polymerase chain reaction (qPCR). RESULTS: STVNa (30 mg/kg) had significant neuroprotective effects 24 hours after pMCAO, including the reduction of the infarct volume and the improvement of the neurological severity score. Immunohistochemistry demonstrated that STVNa significantly increased the number of restored neurons and decreased the number of astrocytes. qPCR also demonstrated that the mRNA expression of inhibitor of nuclear factor kappa-B kinase-α, inhibitor of nuclear factor kappa-B kinase-ß, NF-κB, inhibitor of NF-κB-α, tumor necrosis factor-α, interleukin-1 beta, Bcl2-associated X protein, and caspase-3 were significantly downregulated, whereas B-cell CLL/lymphoma 2 mRNA was upregulated with STVNa treatment compared with vehicle. CONCLUSIONS: These findings demonstrate a neuroprotective role of STVNa during cerebral ischemia, which may result from interactions with the NF-κB signaling pathway and the associated inflammatory and apoptotic responses.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesiones Encefálicas/prevención & control , Diterpenos de Tipo Kaurano/uso terapéutico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , FN-kappa B/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Animales , Lesiones Encefálicas/etiología , Isquemia Encefálica/complicaciones , Caspasa 3/metabolismo , Circulación Cerebrovascular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Examen Neurológico
16.
Biochem Biophys Res Commun ; 477(4): 1045-1050, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27396619

RESUMEN

Cytoplasmic actin and actin-associated proteins have been identified in RABV particles. Although actin is involved in RABV entry into cells, the specific role of actin in RABV budding and release remains unknown. Our study found that RABV M protein-mediated virion budding depends on intact actin filaments. Confocal microscopy demonstrated a block to virions budding, with a number of M protein-mediated budding vesicles detained in the cell cytoplasm. Furthermore, RABV infection resulted in inactivation of cofilin and upregulation of phosphorylated cofilin. Knockdown of cofilin reduced RABV release. These results for the first time indicate that RABV infection resulted in upregulation of phosphorylated cofilin to facililtate actin polymerization for virus budding.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Neuronas/virología , Virus de la Rabia/fisiología , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/metabolismo , Liberación del Virus/fisiología , Animales , Línea Celular , Regulación hacia Abajo/fisiología , Ratones
17.
J Mater Sci Mater Med ; 27(5): 89, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26975746

RESUMEN

To develop adriamycin (ADM)-encapsulated poly(lactic-co-glycolic acid) (PLGA) nanoparticles in a porous nano-hydroxyapatite/collagen scaffold (ADM-PLGA-NHAC). To provide novel strategies for future treatment of osteosarcoma, the properties of the scaffold, including its in vitro extended-release properties, the inhibition effects of ADM-PLGA-NHAC on the osteosarcoma MG63 cells, and its bone repair capacity, were investigated in vivo and in vitro. The PLGA copolymer was utilized as a drug carrier to deliver ADM-PLGA nanoparticles (ADM-PLGA-NP). Porous nano-hydroxyapatite and collagen were used to materials to produce the porous nano-hydroxyapatite/collagen scaffold (NHAC), into which the ADM-PLGA-NP was loaded. The performance of the drug-carrying scaffold was assessed using multiple techniques, including scanning electron microscopy and in vitro extended release. The antineoplastic activities of scaffold extracts on the human osteosarcoma MG63 cell line were evaluated in vitro using the cell counting kit-8 (CCK8) method and live-dead cell staining. The bone repair ability of the scaffold was assessed based on the establishment of a femoral condyle defect model in rabbits. ADM-PLGA-NHAC and NHAC were implanted into the rat muscle bag for immune response experiments. A tumor-bearing nude mice model was created, and the TUNEL and HE staining results were observed under optical microscopy to evaluate the antineoplastic activity and toxic side effects of the scaffold. The composite scaffold demonstrated extraordinary extended-release properties, and its extracts also exhibited significant inhibition of the growth of osteosarcoma MG63 cells. In the bone repair experiment, no significant difference was observed between ADM-PLGA-NHAC and NHAC by itself. In the immune response experiments, ADM-PLGA-NHAC exhibited remarkable biocompatibility. The in vivo antitumor experiment revealed that the implantation of ADM-PLGA-NHAC in the tumor resulted in a improved antineoplastic effect and fewer adverse side effects than direct intraperitoneal injection of ADM. The ADM-PLGA-NHAC developed in this study exhibited excellent extended-release drug properties, bone repairing and antineoplastic efficacy, which make it a promising osteoconductivity material with the capability to inhibit osteosarcoma.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Colágeno/química , Doxorrubicina/química , Durapatita/química , Ácido Láctico/química , Ácido Poliglicólico/química , Andamios del Tejido/química , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Doxorrubicina/farmacología , Femenino , Humanos , Ácido Láctico/farmacología , Masculino , Ratones , Ratones Desnudos , Nanoestructuras , Osteosarcoma , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Conejos , Ratas , Ratas Sprague-Dawley
18.
Biochem Biophys Res Commun ; 464(1): 1-6, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26056009

RESUMEN

Monocyte differentiation into macrophages results in upregulation of miR-29a and scavenger receptor A (SRA) expression, while the expression of RNA binding protein, QKI is suppressed. Since SRA is a functionally important protein in atherosclerosis, it is imperative to understand the various mechanisms involved in its regulation specially the mechanism involving miR-29a. There are individual studies linking miR-29a to SRA or QKI to monocyte differentiation but there is no evidence of any linkage among them. Therefore, we intend to investigate the association among these three, if any, in terms of regulation of SRA expression. Hence, in this study, the differentiated macrophages were initially transfected with miR-29a or its inhibitor and it was shown that QKI is a direct target of mir-29a. In addition, it was also observed by bioinformatics analysis that 3'UTR in SRA mRNA has QKI binding site. So, we attempted to further understand the role of QKI in SRA regulation. The macrophages were manipulated either with overexpression of QKI or by its ablation and it was observed that QKI suppressed SRA at the transcriptional level. Moreover, with the help of luciferase reporter vector, it was shown that QKI inhibited SRA transcription by binding to QRE region in its 3'UTR mRNA. Furthermore, to link the QKI mediated regulation of SRA expression with its functional activity; we analyzed lipid uptake capacity of macrophages transfected with either ectopic OKI plasmid or ablated for QKI. It was observed that, indeed, QKI upregulation inhibits lipid uptake by repressing SRA expression. Overall, our study demonstrates that miR-29a inhibits QKI, which in turn results in upregulation of SRA and lipid uptake.


Asunto(s)
Macrófagos/metabolismo , MicroARNs/genética , Monocitos/metabolismo , Proteínas de Unión al ARN/genética , Receptores Depuradores de Clase A/genética , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Transporte Biológico , Diferenciación Celular , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Metabolismo de los Lípidos , Luciferasas/genética , Luciferasas/metabolismo , Macrófagos/citología , MicroARNs/metabolismo , Datos de Secuencia Molecular , Monocitos/citología , Plásmidos/química , Plásmidos/metabolismo , Cultivo Primario de Células , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Receptores Depuradores de Clase A/metabolismo , Transducción de Señal , Transfección
19.
J Virol ; 87(13): 7608-21, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23637400

RESUMEN

Rabies, as the oldest known infectious disease, remains a serious threat to public health worldwide. The eukaryotic cytosolic chaperonin TRiC/CCT complex facilitates the folding of proteins through ATP hydrolysis. Here, we investigated the expression, cellular localization, and function of neuronal CCTγ during neurotropic rabies virus (RABV) infection using mouse N2a cells as a model. Following RABV infection, 24 altered proteins were identified by using two-dimensional electrophoresis and mass spectrometry, including 20 upregulated proteins and 4 downregulated proteins. In mouse N2a cells infected with RABV or cotransfected with RABV genes encoding nucleoprotein (N) and phosphoprotein (P), confocal microscopy demonstrated that upregulated cellular CCTγ was colocalized with viral proteins N and P, which formed a hollow cricoid inclusion within the region around the nucleus. These inclusions, which correspond to Negri bodies (NBs), did not form in mouse N2a cells only expressing the viral protein N or P. Knockdown of CCTγ by lentivirus-mediated RNA interference led to significant inhibition of RABV replication. These results demonstrate that the complex consisting of viral proteins N and P recruits CCTγ to NBs and identify the chaperonin CCTγ as a host factor that facilitates intracellular RABV replication. This work illustrates how viruses can utilize cellular chaperonins and compartmentalization for their own benefit.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Cuerpos de Inclusión Viral/metabolismo , Neuronas/metabolismo , Virus de la Rabia/fisiología , Rabia/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Cartilla de ADN/genética , Electroforesis en Gel Bidimensional , Técnica del Anticuerpo Fluorescente , Immunoblotting , Inmunoprecipitación , Espectrometría de Masas , Ratones , Microscopía Confocal , Chaperonas Moleculares , Neuronas/virología , Proteínas de la Nucleocápside/metabolismo , Fosfoproteínas/metabolismo , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Estructurales Virales/metabolismo , Replicación Viral/fisiología
20.
Microbiol Immunol ; 58(10): 590-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25082455

RESUMEN

Negri bodies (NBs) are formed in the cytoplasm of rabies virus (RABV)-infected cells and are accompanied by a number of host factors to NBs, in which replication and transcription occur. Here, it was found that chaperonin containing TCP-1 subunit alpha (CCTα) relocalizes to NBs in RABV-infected cells, and that cotransfection of nucleo- and phospho-proteins of RABV is sufficient to recruit CCTα to the NBs' structure. Inhibition of CCTα expression by specific short hairpin RNA knockdown inhibited the replication and transcription of RABV. Therefore, this study showed that the host factor CCTα is associated with RABV infection and is very likely required for efficient virus transcription and replication.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Virus de la Rabia/genética , Rabia/enzimología , Rabia/virología , Transcripción Genética , Replicación Viral , Animales , Línea Celular , Chaperonina con TCP-1/genética , Interacciones Huésped-Patógeno , Humanos , Cuerpos de Inclusión Viral/enzimología , Cuerpos de Inclusión Viral/virología , Ratones , Transporte de Proteínas , Rabia/genética , Virus de la Rabia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA