Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Indoor Air ; 32(1): e12956, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34783390

RESUMEN

Research on individual level polycyclic aromatic hydrocarbons (PAHs) exposure is scarce. Moreover, the independent contribution of ambient- and indoor-origin PAHs to personal exposure remains poorly studied. We performed simultaneous ambient, residential indoor, and personal exposure measurements in a panel of healthy adults to investigate particle-bound PAHs, focusing on their carcinogenic congeners (cPAHs). Average PAH concentrations were much higher in ambient and residential indoor than personal exposure, with distinct seasonal variations. We employed chrysene as a tracer to investigate residential indoor and personal PAHs exposure by origin. Personal cPAH exposure was largely attributable to ambient-origin exposures (95.8%), whereas a considerable proportion of residential indoor PAHs was likely attributable to indoor emissions (33.8%). Benzo[a]pyrene equivalent (BaPeq) concentrations of cPAH accounted for 95.2%-95.6% of total carcinogenic potential. Uncertainties in estimated PAHs (and BaPeq) exposure and cancer risks for adults were calculated using the Monte Carlo simulation. Cancer risks attributable to ambient, residential indoor, and personal cPAH inhalation exposures ranged from 4.0 × 10-6 to 1.0 × 10-5 . A time-activity weighted model was employed for personal PAH exposure estimations. Estimated cPAH exposures demonstrate high cancer risks for adults in Hong Kong, suggesting that exposure to indoor-generated PAHs should be of great concern to the general population.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire Interior , Hidrocarburos Policíclicos Aromáticos , Adulto , Contaminantes Atmosféricos/análisis , Contaminación del Aire Interior/análisis , Monitoreo del Ambiente , Hong Kong , Humanos , Hidrocarburos Policíclicos Aromáticos/análisis , Medición de Riesgo
2.
J Cell Mol Med ; 25(12): 5729-5743, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33949118

RESUMEN

Cyclosporine A (CsA) is an immunosuppressor widely used for the prevention of acute rejection during solid organ transplantation. However, severe nephrotoxicity has substantially limited its long-term usage. Recently, an impaired autophagy pathway was suggested to be involved in the pathogenesis of chronic CsA nephrotoxicity. However, the underlying mechanisms of CsA-induced autophagy blockade in tubular cells remain unclear. In the present study, we observed that CsA suppressed the activation and expression of transcription factor EB (TFEB) by increasing the activation of mTOR, in turn promoting lysosomal dysfunction and autophagy flux blockade in tubular epithelial cells (TECs) in vivo and in vitro. Restoration of TFEB activation by Torin1-mediated mTOR inhibition significantly improved lysosomal function and rescued autophagy pathway activity, suppressing TEC injury. In summary, targeting TFEB-mediated autophagy flux represents a potential therapeutic strategy for CsA-induced nephrotoxicity.


Asunto(s)
Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Ciclosporina/toxicidad , Células Epiteliales/patología , Túbulos Renales/patología , Lisosomas/patología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Inmunosupresores/toxicidad , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Serina-Treonina Quinasas TOR/genética
3.
Lipids Health Dis ; 20(1): 118, 2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34587955

RESUMEN

BACKGROUND: The present study was aimed to establish a prediction model for in-stent restenosis (ISR) in subjects who had undergone percutaneous coronary intervention (PCI) with drug-eluting stents (DESs). MATERIALS AND METHODS: A retrospective cohort study was conducted. From September 2010 to September 2013, we included 968 subjects who had received coronary follow-up angiography after primary PCI. The logistic regression analysis, receiver operator characteristic (ROC) analysis, nomogram analysis, Hosmer-Lemeshow χ2 statistic, and calibration curve were applied to build and evaluate the prediction model. RESULTS: Fifty-six patients (5.79%) occurred ISR. The platelet distribution width (PDW), total cholesterol (TC), systolic blood pressure (SBP), low-density lipoprotein cholesterol (LDL-C), and lesion vessels had significant differences between ISR and non-ISR groups (all P < 0.05). And these variables were independently associated with ISR (all P < 0.05). Furthermore, they were identified as predictors (all AUC > 0.5 and P < 0.05) to establish a prediction model. The prediction model showed a good value of area under curve (AUC) (95%CI): 0.72 (0.64-0.80), and its optimized cut-off was 6.39 with 71% sensitivity and 65% specificity to predict ISR. CONCLUSION: The incidence of ISR is 5.79% in CAD patients with DES implantation in the Xinjiang population, China. The prediction model based on PDW, SBP, TC, LDL-C, and lesion vessels was an effective model to predict ISR in CAD patients with DESs implantation.


Asunto(s)
Plaquetas/metabolismo , Enfermedad de la Arteria Coronaria/sangre , Stents Liberadores de Fármacos/efectos adversos , Lípidos/sangre , Anciano , Angiografía/métodos , Calibración , Enfermedad de la Arteria Coronaria/diagnóstico , Reestenosis Coronaria , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Análisis Multivariante , Nomogramas , Curva ROC , Análisis de Regresión , Estudios Retrospectivos , Riesgo
4.
Environ Res ; 188: 109780, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32554275

RESUMEN

Direct evidence about associations between fine particles (PM2.5) components and the corresponding PM2.5 bioreactivity at the individual level is limited. We conducted a panel study with repeated personal measurements involving 56 healthy residents in Hong Kong. Fractional exhaled nitric oxide (FeNO) levels were measured from these subjects. Out of 56 subjects, 27 (48.2%) participated in concurrent outdoor, indoor, and personal PM2.5 monitoring. Organic carbon (OC), elemental carbon (EC), particle bound-polycyclic aromatic hydrocarbons (PAHs), and phthalates were analyzed. Alteration in cell viability, lactic dehydrogenase (LDH), interleukin-6 (IL-6), and 8-isoprostane by 50 µg/mL PM2.5 extracts was determined in A549 cells in vitro. Moderate heterogeneities were shown in PM2.5 exposures and the corresponding PM2.5 bioreactivity across different sample types. Associations between the analyzed components and PM2.5 bioreactivity were assessed using the multiple regression models. Toxicological results revealed that indoor and personal exposure to OC as well as PAH compounds and their derivatives (e.g., Alkyl-PAHs, Oxy-PAHs) induced cell viability reduction and increase in levels of LDH, IL-6, and 8-isoprostane. Overall, OC in personal exposure played a dominant role in PM2.5-induced bioreactivity. Subsequently, we examined the associations of FeNO with IL-6 and 8-isoprostane levels using mixed-effects models. The results showed that per interquartile change in IL-6 and 8-isoprostane were associated with a 6.4% (p < 0.01) and 11.1% (p < 0.01) increase in FeNO levels, respectively. Our study explored the toxicological properties of chemical components in PM2.5 exposure, which suggested that residential indoors and personal OC and PAHs should be of great concern for human health. These findings indicated that further studies in inflammation and oxidative stress-related illnesses due to particle exposure would benefit from the assessment of in vitro PM2.5 bioreactivity.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire Interior , Hidrocarburos Policíclicos Aromáticos , Contaminantes Atmosféricos/análisis , Contaminantes Atmosféricos/toxicidad , Contaminación del Aire Interior/análisis , Carbono/análisis , Monitoreo del Ambiente , Hong Kong , Humanos , Tamaño de la Partícula , Material Particulado/análisis , Material Particulado/toxicidad , Hidrocarburos Policíclicos Aromáticos/análisis
5.
Med Sci Monit ; 26: e922673, 2020 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-32555132

RESUMEN

BACKGROUND Cell cycle arrest and autophagy have been demonstrated to be involved in various transforming growth factor (TGF)-ß-mediated phenotype alterations of tubular epithelial cells (TECs) and tubulointerstitial fibrosis. But the relationship between cell cycle arrest and the autophagy induced by TGF-ß has not been explored well. MATERIAL AND METHODS The effects of autophagy inhibition on TGF-ß-induced cell cycle arrest in TECs were explored in vitro. Human kidney-2 (HK-2) cells were stimulated by TGF-ß with or without a combined treatment of autophagy inhibitor chloroquine (CQ) or bafilomycin A1 (Baf). RESULTS Autophagy inhibition by CQ or Baf promotes the suppression of growth in TGF-ß-treated HK-2 cells, as detected by the Cell Counting Kit-8 (CCK-8) method. In addition, CQ or Baf stimulation enhances G1 arrest in TGF-ß treated HK-2 cells, as investigated using propidium iodide (PI) staining and flow cytometry, which was further confirmed by a decrease in the expression of phosphorylated retinoblastoma protein (p-RB) and cyclin-dependent kinase 4 (CDK4). The upregulation of p21 induced by CQ or Baf may mediate an enhanced G1 arrest in TGF-ß treated HK-2 cells. Western blot analysis showed that TGF-ß-induced expression of extracellular matrix fibronectin was notably upregulated in the presence of autophagy inhibitors. CONCLUSIONS Inhibition of autophagy sensitizes the TECs to G1 arrest and proliferation suppression induced by TGF-ß that contributes to the induction of tubulointerstitial fibrosis.


Asunto(s)
Autofagia/efectos de los fármacos , Cloroquina/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Macrólidos/farmacología , Insuficiencia Renal Crónica/patología , Factor de Crecimiento Transformador beta/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibronectinas/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Humanos , Técnicas In Vitro , Túbulos Renales/citología , Insuficiencia Renal Crónica/metabolismo , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo
6.
Lipids Health Dis ; 19(1): 150, 2020 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-32580730

RESUMEN

BACKGROUND: Inflammation and oxidative stress play predominant roles in the initiation and progression of ischaemia/reperfusion (I/R) injury, with nuclear factor kappa B (NF-κB) serving as a crucial mediator. Overexpression of the inhibitor of κB alpha (IκBα) gene is hypothesized to have protective effects against apoptosis and autophagy in cardiomyocytes subjected to hydrogen peroxide (H2O2) by inhibiting the NF-κB pathway. METHODS: The IκBαS32A, S36A gene was transfected via adeno-associated virus serotype 9 (AAV9) delivery into neonatal rat ventricular cardiomyocytes (NRVMs) prior to H2O2 treatment. NRVMs were divided into control, H2O2, GFP + H2O2, IκBα+H2O2, and pyrrolidine dithiocarbamate (PDTC) + H2O2 groups. Nuclear translocation of the NF-κB p65 subunit was evaluated by immunofluorescence and Western blotting. Cell viability was assessed by Cell Counting Kit-8 assay. Supernatant lactate dehydrogenase (LDH) and intracellular malondialdehyde (MDA) were measured to identify H2O2-stimulated cytotoxicity. Apoptosis was determined by Annexin V-PE/7-AAD staining, and the mitochondrial membrane potential (ΔΨm) was detected by JC-1 staining. Western blotting was used to detect apoptosis- and autophagy-related proteins. RESULTS: IκBα transfection significantly increased cell viability and ΔΨm but decreased the supernatant LDH and cellular MDA levels in cardiomyocytes exposed to H2O2. Meanwhile, IκBα overexpression decreased H2O2-induced apoptosis by upregulating the Bcl-2/Bax ratio and reduced autophagy by downregulating the expression of Beclin-1 and the LC3-II/LC3-I ratio. These effects partly accounted for the ability of IκBα to inhibit the NF-κB signalling pathway, as evidenced by decreases in p65 phosphorylation and nuclear translocation. Indeed, the effects of inactivation of NF-κB signalling with the specific inhibitor PDTC resembled the cardioprotective effects of IκBα during H2O2 stimulation. CONCLUSION: IκBα overexpression can ameliorate H2O2-induced apoptosis, autophagy, oxidative injury, and ΔΨm loss through inhibition of the NF-κB signalling pathway. These findings suggest that IκBα transfection can result in successful resistance to oxidative stress-induced damage by inhibiting NF-κB activation, which may provide a potential therapeutic target for the prevention of myocardial I/R injury.


Asunto(s)
Peróxido de Hidrógeno/farmacología , Miocitos Cardíacos/patología , Inhibidor NF-kappaB alfa/genética , FN-kappa B/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Células Cultivadas , Dependovirus/genética , Regulación de la Expresión Génica , Peróxido de Hidrógeno/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Pirrolidinas/farmacología , Ratas Sprague-Dawley , Tiocarbamatos/farmacología , Transfección
7.
Biochem Biophys Res Commun ; 512(1): 125-130, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30876692

RESUMEN

Oxidative stress injury is one of the main mechanisms of ischemia-reperfusion (I/R) injury. The extracellular signal-regulated kinase (ERK1/2) pathway plays an important role in cardioprotective during acute myocardial infarction. In this study, we used constitutively active MEK1 gene (CaMEK) transfection strategy to investigate whether CaMEK provides a protective effect against apoptosis and autophagy induced by Hydrogen peroxide (H2O2) in neonatal rat cardiac ventricular cardiomyocytes (NCMs) and the underlying mechanisms. As a result, CaMEK attenuated H2O2-induced apoptosis and cytotoxicity in NCMs, evidenced by decreased apoptotic cells and the ratio of Bax/Bcl-2, increased the mitochondrial membrane potential (Δψm) and cell vitality and reduced the level of lactate dehydrogenase (LDH). Further studies revealed that CaMEK attenuated H2O2-induced autophagy, evidenced by the decreased LC3-Ⅱ/LC3-Ⅰratio and SQSTM1/p62 (p62) degradation. Furthermore, we demonstrated that CaMEK phosphorylated the ERK1/2 pathway-related proteins, ERK1/2, p70S6K and GSK3ß, in NCMs with H2O2 stimulation. In contrast, these effects could be reversed by co-treatment with the ERK1/2 inhibitor, PD98059. These results suggest that CaMEK plays an important role in protecting cardiomyocytes against H2O2-induced injury and autophagy in NCMs via ERK1/2 pathway. Therefore, transfection of CaMEK may provide a hopeful therapeutic strategy for I/R.


Asunto(s)
MAP Quinasa Quinasa 1/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Cardiotónicos/metabolismo , Células Cultivadas , Femenino , Peróxido de Hidrógeno/toxicidad , MAP Quinasa Quinasa 1/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
8.
Clin Sci (Lond) ; 133(5): 665-680, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30804219

RESUMEN

Ischemic preconditioning (IPC) is an endogenous protection strategy against myocardial ischemia-reperfusion (I/R) injury. Macrophage migration inhibitory factor (MIF) released from the myocardium subjected to brief periods of ischemia confers cardioprotection. We hypothesized that MIF plays an essential role in IPC-induced cardioprotection. I/R was induced either ex vivo or in vivo in male wild-type (WT) and MIF knockout (MIFKO) mice with or without proceeding IPC (three cycles of 5-min ischemia and 5-min reperfusion). Indices of myocardial injury, regional inflammation and cardiac function were determined to evaluate the extent of I/R injury. Activations of the reperfusion injury salvage kinase (RISK) pathway, AMP-activated protein kinase (AMPK) and their downstream components were investigated to explore the underlying mechanisms. IPC conferred prominent protection in WT hearts evidenced by reduced infarct size (by 33-35%), myocyte apoptosis and enzymatic markers of tissue injury, ROS production, inflammatory cell infiltration and MCP1/CCR2 expression (all P<0.05). IPC also ameliorated cardiac dysfunction both ex vivo and in vivo These protective effects were abolished in MIFKO hearts. Notably, IPC mediated further activations of RISK pathway, AMPK and the membrane translocation of GLUT4 in WT hearts. Deletion of MIF blunted these changes in response to IPC, which is the likely basis for the absence of protective effects of IPC against I/R injury. In conclusion, MIF plays a critical role in IPC-mediated cardioprotection under ischemic stress by activating RISK signaling pathway and AMPK. These results provide an insight for developing a novel therapeutic strategy that target MIF to protect ischemic hearts.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Precondicionamiento Isquémico Miocárdico/métodos , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR2/metabolismo , Transducción de Señal , Remodelación Ventricular
9.
Lipids Health Dis ; 18(1): 159, 2019 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391051

RESUMEN

BACKGROUND: The study was designed to investigate lipid profile and SYNTAX score in patients with non-ST segment elevation myocardial infarction (NSTEMI). METHODS: 311 patients with NSTEMI were enrolled. The demographic, clinical data, blood samples and SYNTAX score were documented. The Pearson linear correlation was used to detect confounding factors linearly correlated with SYNTAX score. The significantly correlated confounding factors were put into the multiple linear regressions. RESULTS: The Pearson linear correlation showed that high-density lipoprotein- cholesterol (HDL-C) and apolipoprotein A1 (ApoA1) were significantly correlated with Syntax Score (r = - 0.119, P = 0.044 and r = - 0.182, P = 0.002, respectively). The multiple linear regressions for Syntax Score were built using HDL-C and ApoA1, respectively. After the adjustment of other significantly correlated confounding factors such as white blood cell count (WBC), myohemoglobin (MB), glutamic-oxalacetic transaminase (AST) and creatinine, the ApoA1 still showed significant association with Syntax Score (ß = - 0.151, P = 0.028). The area under curve was (AUC) 0.624 and the optimal cutoff value is 1.07 g/L when using ApoA1 to predict moderate and severe coronary artery lesions. The patients with ApoA1 ≥ 1.07 g/L and < 1.07 g/L have the Syntax Scores of 12.21 ± 11.58 and 16.33 ± 11.53, respectively (P = 0.001). CONCLUSIONS: The ApoA1 is the only lipid factor significantly associated with complexity of coronary artery lesion in patients with NSTEMI, the patients with ApoA1 < 1.07 g/L may have more complex coronary artery lesions.


Asunto(s)
Apolipoproteína A-I/sangre , Infarto del Miocardio sin Elevación del ST/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Aspartato Aminotransferasas/sangre , Biomarcadores/sangre , HDL-Colesterol/sangre , Creatinina/sangre , Estudios Transversales , Femenino , Humanos , Recuento de Leucocitos , Modelos Lineales , Masculino , Persona de Mediana Edad , Mioglobina/sangre , Infarto del Miocardio sin Elevación del ST/sangre , Infarto del Miocardio sin Elevación del ST/fisiopatología , Índice de Severidad de la Enfermedad
10.
Environ Res ; 164: 24-31, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29462750

RESUMEN

BACKGROUND: Given the lack of research on the personal exposure to fine particles (PM2.5) in Hong Kong, we examined the association between short-term personal exposure to PM2.5 and their constituents and inflammation in exhaled breath in a sample of healthy adult residents. METHOD: Forty-six participants underwent personal PM2.5 monitoring for averagely 6 days to obtain 276 samples. Fractional exhaled nitric oxide (FeNO), a biomarker of inflammation in exhaled breath, was measured at the end of each 24-h personal monitoring. PM2.5 chemical constituents, including organic carbon, elemental carbon, 16 polycyclic aromatic hydrocarbons (PAHs), and 6 phthalate esters, were speciated from the personal samples collected. A mixed-effects model was used to estimate the association of PM2.5 and their constituents with FeNO. The comparison was also made with parallel analyses using ambient concentrations. RESULTS: Personal exposures to PM2.5 (28.1 ±â€¯23.3 µg/m3) were higher than the ambient levels (13.3 ±â€¯6.4 µg/m3) monitored by stations. The composition profile and personal-to-ambient concentration ratio varied among subjects with different occupations. An interquartile range (IQR) change in personal exposure to PM2.5 was positively associated with 12.8% increase in FeNO (95% confidence interval, CI: 5.5-20.7%), while nil association was found for ambient PM2.5. Among the constituents measured, only the carcinogenic PAHs were significantly associated with 12% increase in FeNO responses (95% CI, 0.0-25.6%). CONCLUSION: In conclusion, our study provides the first understanding about personal exposure to PM2.5 and possible sources in Hong Kong. The results also showed that personal exposure to PM2.5 and c-PAHs were linked to increased FeNO levels among healthy adults.


Asunto(s)
Contaminantes Atmosféricos , Hidrocarburos Policíclicos Aromáticos , Adulto , Contaminantes Atmosféricos/análisis , Hong Kong , Humanos , Inflamación , Material Particulado/análisis
11.
J Mol Cell Cardiol ; 103: 56-64, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28088561

RESUMEN

Nuclear factor κappa B (NF-κB) is an important transcription factor in the development and progression of coronary artery disease (CAD). Recent evidence suggests that -94 ATTG ins/del mutant in the promoter of NFKB1 gene is an essential functional mutant. The present study demonstrated the frequencies of the del/del (DD) genotype and del (D) allele were significantly higher in CAD patients than in controls. CAD patients carrying mutant DD genotype had worse stenosis of diseased coronary arteries compared to those carrying ins/ins (II) or ins/del (ID) genotype. Plasma levels of endothelial nitric oxide synthase (eNOS) were lower, while inflammatory cytokine incnterlukin-6 (IL-6) was higher in CAD patients with DD genotype than those with II or ID genotype (both P<0.05). In vitro study showed that mutant human umbilical vein endothelial cells (DD genotype HUVECs) were more susceptible to H2O2-induced apoptosis, which was accompanied with a decreased Bcl-2 expression. Further, mutant HUVECs had lower eNOS but higher IL-6 mRNA levels and decreased phosphorylation of eNOS under H2O2-stimulation (both P<0.05). Compared to wild type cells (II genotype), significantly downregulated protein expression of total NF-κB p50 subunit were observed in mutant HUVECs with or without oxidative stress, and a lower expression of unclear p50 was associated with a decreased p50 nuclear translocation in mutant HUVECs versus wild type cells under H2O2-stimulation (both P<0.05). In conclusion, mutant DD genotype of NFKB1 gene is associated with the risk and severity of CAD. Dwonregulation of NF-κB p50 subunit leads to exacerbated endothelial dysfunction and apoptosis and enhanced inflammatory response that is the potential underlying mechanism.


Asunto(s)
Enfermedad de la Arteria Coronaria/diagnóstico , Enfermedad de la Arteria Coronaria/genética , Predisposición Genética a la Enfermedad , Genotipo , Mutación , Subunidad p50 de NF-kappa B/genética , Anciano , Alelos , Apoptosis/efectos de los fármacos , Biomarcadores , Enfermedad de la Arteria Coronaria/metabolismo , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Femenino , Expresión Génica , Estudios de Asociación Genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Masculino , Persona de Mediana Edad , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Riesgo , Índice de Severidad de la Enfermedad
12.
Clin Exp Pharmacol Physiol ; 42(10): 1108-17, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26173818

RESUMEN

Adeno-associated virus (AAV) has become one of the most promising gene transfer tools for gene therapy. This work aims to evaluate tropism, gene transfer efficiency and safety of AAV9 vectors produced with recombinant baculovirus (rBac)-based system. AAV9-CMV-GFP and AAV9-CBA-GFP were produced using a rBac system, 1 × 10(11) particles of each vectors were administered intravenously (i.v.) into mice and animals were killed at 1, 2, 3, 4, 5 and 8 weeks after administration. The GFP expression in different organs was analyzed by fluorescence imaging and Western blot. Viral genomic quantities were measured using qPCR. In vitro transduction efficiency of AAV9 vectors in primary cardiomyocytes and hepatocytes was determined by flow cytometry. Toxicity of AAV9 vectors was evaluated by determining certain cardiac and liver injury biomarkers and renal function test in vivo and TUNEL analysis in vitro. The data showed that AAV9 viral particles packaged by the rBac system were fully functional in vivo and in vitro. The CMV promoter predominantly induced higher cardiac GFP transgene expression and DNA copy numbers while the CBA promoter resulted in robust GFP expression and high vector DNA copy numbers in mouse liver, both in a time-dependent increased manner. Such distinct preferential effects were also observed in the heart and liver as early as 3 and 5 days after co-infection. Both the AAV9-CMV and AAV9-CBA viral packages did not induce heart, liver and renal damage and cell apoptosis. These results indicated that AAV9-CMV can efficiently and safely direct cardiac gene transfer, whereas AAV9-CBA is preferential for liver gene delivery.


Asunto(s)
Dependovirus/genética , Hepatocitos/metabolismo , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas/genética , Transducción Genética/métodos , Transgenes/genética , Actinas/genética , Animales , Apoptosis/genética , Pollos/genética , Citomegalovirus/genética , Dependovirus/fisiología , Vectores Genéticos/genética , Hepatocitos/citología , Hepatocitos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/citología , Miocitos Cardíacos/virología , Seguridad , Tropismo Viral
13.
Pharmaceuticals (Basel) ; 17(5)2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38794195

RESUMEN

Chronic kidney disease (CKD) affects more than 10% of the global population, and its incidence is increasing, partially due to an increase in the prevalence of disease risk factors. Acute kidney injury (AKI) is an independent risk factor for CKD and end-stage renal disease (ESRD). The pathogenic mechanisms of CKD provide several potential targets for its treatment. However, due to off-target effects, conventional drugs for CKD typically require high doses to achieve adequate therapeutic effects, leading to long-term organ toxicity. Therefore, ideal treatments that completely cure the different types of kidney disease are rarely available. Several approaches for the drug targeting of the kidneys have been explored in drug delivery system research. Nanotechnology-based drug delivery systems have multiple merits, including good biocompatibility, suitable degradability, the ability to target lesion sites, and fewer non-specific systemic effects. In this review, the development, potential, and limitations of low-molecular-weight protein-lysozymes, polymer nanomaterials, and lipid-based nanocarriers as drug delivery platforms for treating AKI and CKD are summarized.

14.
Front Physiol ; 14: 1187207, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37256065

RESUMEN

Peritoneal dialysis (PD) is a widely accepted renal replacement therapy for patients with end-stage renal disease (ESRD). Morphological and functional changes occur in the peritoneal membranes (PMs) of patients undergoing long-term PD. Peritoneal fibrosis (PF) is a common PD-related complication that ultimately leads to PM injury and peritoneal ultrafiltration failure. Autophagy is a cellular process of "self-eating" wherein damaged organelles, protein aggregates, and pathogenic microbes are degraded to maintain intracellular environment homeostasis and cell survival. Growing evidence shows that autophagy is involved in fibrosis progression, including renal fibrosis and hepatic fibrosis, in various organs. Multiple risk factors, including high-glucose peritoneal dialysis solution (HGPDS), stimulate the activation of autophagy, which participates in PF progression, in human peritoneal mesothelial cells (HPMCs). Nevertheless, the underlying roles and mechanisms of autophagy in PF progression remain unclear. In this review, we discuss the key roles and potential mechanisms of autophagy in PF to offer novel perspectives on future therapy strategies for PF and their limitations.

15.
Cell Death Dis ; 14(7): 473, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37500613

RESUMEN

The endoplasmic reticulum (ER) plays important roles in biosynthetic and metabolic processes, including protein and lipid synthesis, Ca2+ homeostasis regulation, and subcellular organelle crosstalk. Dysregulation of ER homeostasis can cause toxic protein accumulation, lipid accumulation, and Ca2+ homeostasis disturbance, leading to cell injury and even death. Accumulating evidence indicates that the dysregulation of ER homeostasis promotes the onset and progression of kidney diseases. However, maintaining ER homeostasis through unfolded protein response, ER-associated protein degradation, autophagy or ER-phagy, and crosstalk with other organelles may be potential therapeutic strategies for kidney disorders. In this review, we summarize the recent research progress on the relationship and molecular mechanisms of ER dysfunction in kidney pathologies. In addition, the endogenous protective strategies for ER homeostasis and their potential application for kidney diseases have been discussed.


Asunto(s)
Estrés del Retículo Endoplásmico , Enfermedades Renales , Humanos , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Respuesta de Proteína Desplegada , Enfermedades Renales/patología , Autofagia , Homeostasis , Lípidos
16.
Biomed Pharmacother ; 165: 115122, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37413899

RESUMEN

Nephrotoxicity is a major side effect of cisplatin treatment of solid tumors in the clinical setting. Long-term low-dose cisplatin administration causes renal fibrosis and inflammation. However, few specific medicines with clinical application value have been developed to reduce or treat the nephrotoxic side effects of cisplatin without affecting its tumor-killing effect. The present study analyzed the potential reno-protective effect and mechanism of asiatic acid (AA) in long-term cisplatin-treated nude mice suffering from tumors. AA treatment significantly attenuated renal injury, inflammation, and fibrosis induced by long-term cisplatin injection in tumor-bearing mice. AA administration notably suppressed tubular necroptosis and improved the autophagy-lysosome pathway disruption caused by chronic cisplatin treatment in tumor-transplanted nude mice and HK-2 cells. AA promoted transcription factor EB (TFEB)-mediated lysosome biogenesis and reduced the accumulation of damaged lysosomes, resulting in enhanced autophagy flux. Mechanistically, AA increased TFEB expression by rebalancing Smad7/Smad3, whereas siRNA inhibition of Smad7 or TFEB abolished the effect of AA on autophagy flux in HK-2 cells. In addition, AA treatment did not weaken, but actually enhanced the anti-tumor effect of cisplatin, as evidenced by the promoted tumor apoptosis and inhibited proliferation in nude mice. In summary, AA alleviates cisplatin-induced renal fibrosis in tumor-bearing mice by improving the TFEB-mediated autophagy-lysosome pathway.


Asunto(s)
Cisplatino , Neoplasias , Ratones , Animales , Cisplatino/farmacología , Ratones Desnudos , Autofagia , Fibrosis , Neoplasias/metabolismo , Inflamación/metabolismo , Lisosomas/metabolismo
17.
J Immunol Res ; 2023: 7625817, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37692838

RESUMEN

Systemic lupus erythematosus (SLE) is a complex autoimmune disease. Approximately one-third to two-thirds of the patients with SLE progress to lupus nephritis (LN). The pathogenesis of SLE and LN has not yet been fully elucidated, and effective treatment for both conditions is lacking. The endoplasmic reticulum (ER) is the largest intracellular organelle and is a site of protein synthesis, lipid metabolism, and calcium storage. Under stress, the function of ER is disrupted, and the accumulation of unfolded or misfolded proteins occurs in ER, resulting in an ER stress (ERS) response. ERS is involved in the dysfunction of B cells, macrophages, T cells, dendritic cells, neutrophils, and other immune cells, causing immune system disorders, such as SLE. In addition, ERS is also involved in renal resident cell injury and contributes to the progression of LN. The molecular chaperones, autophagy, and proteasome degradation pathways inhibit ERS and restore ER homeostasis to improve the dysfunction of immune cells and renal resident cell injury. This may be a therapeutic strategy for SLE and LN. In this review, we summarize advances in this field.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Humanos , Nefritis Lúpica/terapia , Lupus Eritematoso Sistémico/terapia , Riñón , Estrés del Retículo Endoplásmico , Autofagia
18.
Theranostics ; 12(1): 379-395, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34987651

RESUMEN

Rationale: Poor ß cell proliferation is one of the detrimental factors hindering islet cell replacement therapy for patients with diabetes. Smad3 is an important transcriptional factor of TGF-ß signaling and has been shown to promote diabetes by inhibiting ß cell proliferation. Therefore, we hypothesize that Smad3-deficient islets may be a novel cell replacement therapy for diabetes. Methods: We examined this hypothesis in streptozocin-induced type-1 diabetic mice and type-2 diabetic db/db mice by transplanting Smad3 knockout (KO) and wild type (WT) islets under the renal capsule, respectively. The effects of Smad3KO versus WT islet replacement therapy on diabetes and diabetic kidney injury were examined. In addition, RNA-seq was applied to identify the downstream target gene underlying Smad3-regulated ß cell proliferation in Smad3KO-db/db versus Smad3WT-db/db mouse islets. Results: Compared to Smad3WT islet therapy, treatment with Smad3KO islets produced a much better therapeutic effect on both type-1 and type-2 diabetes by significantly lowering serum levels of blood glucose and HbA1c and protected against diabetic kidney injuries by preventing an increase in serum creatinine and the development of proteinuria, mesangial matrix expansion, and fibrosis. These were associated with a significant increase in grafted ß cell proliferation and blood insulin levels, resulting in improved glucose intolerance. Mechanistically, RNA-seq revealed that compared with Smad3WT-db/db mouse islets, deletion of Smad3 from db/db mouse islets markedly upregulated E2F3, a pivotal regulator of cell cycle G1/S entry. Further studies found that Smad3 could bind to the promoter of E2F3, and thus inhibit ß cell proliferation via an E2F3-dependent mechanism as silencing E2F3 abrogated the proliferative effect on Smad3KO ß cells. Conclusion: Smad3-deficient islet replacement therapy can significantly improve both type-1 and type-2 diabetes and protect against diabetic kidney injury, which is mediated by a novel mechanism of E2F3-dependent ß cell proliferation.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Factor de Transcripción E2F3/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína smad3/metabolismo , Animales , Células Secretoras de Insulina/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Eur J Med Res ; 27(1): 176, 2022 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-36088368

RESUMEN

Hyperuricemia can induce acute and chronic kidney damage, but the pathological mechanism remains unclear. The potential role of AMP-activated protein kinase (AMPK) α2 in hyperuricemia-induced renal injury was investigated in this study. Acute and chronic hyperuricemic nephropathy was induced by administering intraperitoneal injections of uric acid and oxonic acid to AMPK α2 knockout and wild-type mice. Changes in renal function, histopathology, inflammatory cell infiltration, renal interstitial fibrosis, and urate deposition were analyzed. In both acute and chronic hyperuricemic nephropathy mouse models, knockout of AMPK α2 significantly reduced serum creatinine levels and renal pathological changes. The tubular expression of kidney injury molecule-1 was also reduced in hyperuricemic nephropathy mice deficient in AMPK α2. In addition, knockout of AMPK α2 significantly suppressed the infiltration of renal macrophages and progression of renal interstitial fibrosis in mice with chronic hyperuricemic nephropathy. Knockout of AMPK α2 reduced renal urate crystal deposition, probably through increasing the expression of the uric acid transporter, multidrug resistance protein 4. In summary, AMPK α2 is involved in acute and chronic hyperuricemia-induced kidney injury and may be associated with increased urate crystal deposition in the kidney.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Hiperuricemia , Enfermedades Renales , Fallo Renal Crónico , Proteínas Quinasas Activadas por AMP/genética , Animales , Modelos Animales de Enfermedad , Fibrosis , Hiperuricemia/inducido químicamente , Hiperuricemia/genética , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Ratones , Ratones Noqueados , Ácido Úrico/efectos adversos , Ácido Úrico/metabolismo
20.
Lupus Sci Med ; 9(1)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35414608

RESUMEN

OBJECTIVE: Lupus nephritis (LN) is a major complication and cause of death among patients with SLE. This research used in vivo and in vitro experiments to explore the therapeutic potential of metformin in kidney injury from LN-induced inflammation. METHODS: In vivo study, 8-week-old MRL/MpJ-Faslpr/J (MRL/lpr) mice were randomly divided into two groups (n=12 each): daily administration of 0.3 mg/mL metformin in drinking water and control (water only). Body weight and urinary samples were measured biweekly. Mice were sacrificed after 8-week treatment to harvest serum, lymph nodes, spleen and kidneys. In vitro study, human kidney-2 (HK-2) cells were pretreated with 1 mM metformin for 1 hour and then stimulated with 20 µg/mL lipopolysaccharides (LPS) or 10 ng/mL tumour necrosis factor-α (TNF-α) for another 48 hours. Protein was collected for subsequent analysis. RESULTS: We found that metformin administration improved renal function in MRL/lpr lupus-prone mice, measured by decreased urea nitrogen and urinary proteins. Metformin reduced immunoglobulin G and complement C3 deposition in glomeruli. The treatment also downregulated systemic and renal inflammation, as seen in decreased renal infiltration of F4/80-positive macrophages and reduced splenic and renal MCP-1 (monocyte chemoattractant protein-1) and TNF-α, and renal IL-1ß (interleukin 1ß) expression. Metformin administration decreased renal expression of necroptosis markers p-RIPK1 (phosphorylated receptor-interacting protein kinase 1) and p-MLKL, along with tubular injury marker KIM-1 (kidney injury molecule-1) in lupus mice. In addition, metformin alleviated the necroptosis of HK-2 cells stimulated by LPS and TNF-α, evidencing by a decrease in the expression of necroptosis markers p-RIPK1, p-RIPK3 and p-MLKL, and the inflammasome-related markers NLRP3 (NLR family pyrin domain containing 3), ASC (apoptosis-associated speck-like protein containing a CARD), caspase-1. Mechanistically, metformin treatment upregulated p-AMPK (phosphorylated AMP-activated protein kinase) and downregulated p-STAT3 (phosphorylated signal transducer and activator of transcription 3) expression in the kidneys. Moreover, AMPKα2 knockdown abolished the protective effects of metformin in vitro. CONCLUSIONS: Metformin alleviated kidney injury in LN though suppressing renal necroptosis and inflammation via the AMPK/STAT3 pathway.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Metformina , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/farmacología , Animales , Humanos , Inflamación , Riñón/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/uso terapéutico , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/tratamiento farmacológico , Nefritis Lúpica/complicaciones , Nefritis Lúpica/tratamiento farmacológico , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Ratones Endogámicos MRL lpr , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/farmacología , Factor de Transcripción STAT3/uso terapéutico , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA