Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 9(7)2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38421727

RESUMEN

Myocardial ischemia/reperfusion (MI/R) injury is a major cause of adverse outcomes of revascularization following myocardial infarction. Anaerobic glycolysis during myocardial ischemia is well studied, but the role of aerobic glycolysis during the early phase of reperfusion is incompletely understood. Lactylation of Histone H3 (H3) is an epigenetic indicator of the glycolytic switch. Heat shock protein A12A (HSPA12A) is an atypic member of the HSP70 family. In the present study, we report that, during reperfusion following myocardial ischemia, HSPA12A was downregulated and aerobic glycolytic flux was decreased in cardiomyocytes. Notably, HSPA12A KO in mice exacerbated MI/R-induced aerobic glycolysis decrease, cardiomyocyte death, and cardiac dysfunction. Gain- and loss-of-function studies demonstrated that HSPA12A was required to support cardiomyocyte survival upon hypoxia/reoxygenation (H/R) challenge and that its protective effects were mediated by maintaining aerobic glycolytic homeostasis for H3 lactylation. Further analyses revealed that HSPA12A increased Smurf1-mediated Hif1α protein stability, thus increasing glycolytic gene expression to maintain appropriate aerobic glycolytic activity to sustain H3 lactylation during reperfusion and, ultimately, improving cardiomyocyte survival to attenuate MI/R injury.


Asunto(s)
Infarto del Miocardio , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , Animales , Ratones , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Infarto del Miocardio/metabolismo , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo
2.
Small ; 19(52): e2304427, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37653590

RESUMEN

As an ideal in vitro model, brain-on-chip (BoC) is an important tool to comprehensively elucidate brain characteristics. However, the in vitro model for the definition scope of BoC has not been universally recognized. In this review, BoC is divided into brain cells-on-a- chip, brain slices-on-a-chip, and brain organoids-on-a-chip according to the type of culture on the chip. Although these three microfluidic BoCs are constructed in different ways, they all use microfluidic chips as carrier tools. This method can better meet the needs of maintaining high culture activity on a chip for a long time. Moreover, BoC has successfully integrated cell biology, the biological material platform technology of microenvironment on a chip, manufacturing technology, online detection technology on a chip, and so on, enabling the chip to present structural diversity and high compatibility to meet different experimental needs and expand the scope of applications. Here, the relevant core technologies, challenges, and future development trends of BoC are summarized.


Asunto(s)
Encéfalo , Microfluídica , Microfluídica/métodos , Organoides , Dispositivos Laboratorio en un Chip
3.
Diagnostics (Basel) ; 13(11)2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37296823

RESUMEN

Deep learning, with continuous development, has achieved relatively good results in the field of left atrial segmentation, and numerous semi-supervised methods in this field have been implemented based on consistency regularization to obtain high-performance 3D models by training. However, most semi-supervised methods focus on inter-model consistency and ignore inter-model discrepancy. Therefore, we designed an improved double-teacher framework with discrepancy information. Herein, one teacher learns 2D information, another learns both 2D and 3D information, and the two models jointly guide the student model for learning. Simultaneously, we extract the isomorphic/heterogeneous discrepancy information between the predictions of the student and teacher model to optimize the whole framework. Unlike other semi-supervised methods based on 3D models, ours only uses 3D information to assist 2D models, and does not have a fully 3D model, thus addressing the large memory consumption and limited training data of 3D models to some extent. Our approach shows excellent performance on the left atrium (LA) dataset, similar to that of the best performing 3D semi-supervised methods available, compared to existing techniques.

4.
BioDrugs ; 37(1): 89-98, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36417156

RESUMEN

OBJECTIVE: We aimed to evaluate the similarity of BAT2206 to its originator, ustekinumab, including pharmacokinetic profiles, immunogenicity, and safety in healthy Chinese male subjects. METHODS: This was a double-blinded, randomized, single-dose, parallel-group clinical trial, in which 270 healthy male subjects were enrolled to receive a single subcutaneous injection (45 mg) of either BAT2206 or ustekinumab (European Union or USA) at a 1:1:1 ratio. The pairwise pharmacokinetic similarities and the safety and immunogenicity of both drugs were evaluated and compared. RESULTS: The results showed that the 90% confidence interval of the geometric mean ratio for primary pharmacokinetic parameters (maximum plasma concentration and area under the plasma concentration-time curve from time zero to infinity) among BAT2206 and ustekinumab (USA or European Union sourced) groups were all within the predefined equivalent interval of 80-125%. Furthermore, all the groups had similar incidences of treatment-emergent adverse events, in which the majority of cases belonged to Common Terminology Criteria for the Classification of Adverse Events Grade 1 or 2. Anti-drug antibodies were detected in 54 (20.1%) subjects, namely 24 (26.7%), 13 (14.8%), and 17 (18.9%) patients in the BAT2206, ustekinumab (European Union), and ustekinumab (USA) groups, respectively. In contrast, the incidences of positive neutralizing antibodies were similar among the three groups. CONCLUSIONS: Pharmacokinetic similarity between BAT2206 and ustekinumab (USA or European Union sourced) was confirmed. The three groups had similar safety profiles, and the investigational drugs were well tolerated by subjects. CLINICAL TRIAL REGISTRATION: This study was registered with ClinicalTrials.gov (NCT04371185).


Asunto(s)
Biosimilares Farmacéuticos , Pueblos del Este de Asia , Ustekinumab , Humanos , Masculino , Biosimilares Farmacéuticos/efectos adversos , Biosimilares Farmacéuticos/farmacocinética , Método Doble Ciego , Voluntarios Sanos , Ustekinumab/efectos adversos , Ustekinumab/inmunología , Ustekinumab/farmacocinética
5.
Expert Opin Investig Drugs ; 30(1): 77-83, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33241960

RESUMEN

Background: To compare the pharmacokinetic (PK) profile, safety, and immunogenicity between golimumab and the biosimilar BAT2506 in healthy Chinese male subjects. Research design and methods: A total of 180 healthy male subjects were recruited for this randomized, double-blinded, single-dose, parallel study. They received 50 mg BAT2506 or golimumab (1:1 ratio) by single subcutaneous injection. The evaluation index included maximum plasma concentration (Cmax), area under the plasma concentration-time curve (AUC0-t, AUC0-∞), safety, and immunogenicity. Results: The results showed that the 90% confidence interval (CI) of the geometric mean ratio (GMR) of BAT2506 to reference drug (golimumab) for Cmax, AUC0-∞ and AUC0-t were 99.26% (90.59-108.76%), 102.06% (93.31%-111.64%), and 102.05% (93.51-111.38%), respectively. All 90% CIs were within the range of 80-125% range, which is the limitation of the equivalence margin. Furthermore, similarity of treatment-emergent adverse events was also found between the two drugs. A total of 14 subjects (7.8%) developed anti-drug antibody after administration. Conclusions: Our study confirmed the PK similarity between BAT2506 and golimumab, and showed good tolerance of BAT2506 in healthy subjects. There were no differences in safety and immunogenicity between the two drugs. Therefore, BAT2506 meets the criteria for biosimilarity to golimumab. Trial registration: The trial is registered at ClinicalTrials.gov (CT.gov identifier: NCT04152759).


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Biosimilares Farmacéuticos/administración & dosificación , Inhibidores del Factor de Necrosis Tumoral/administración & dosificación , Adulto , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Área Bajo la Curva , Pueblo Asiatico , Biosimilares Farmacéuticos/efectos adversos , Biosimilares Farmacéuticos/farmacocinética , Método Doble Ciego , Humanos , Inyecciones Subcutáneas , Masculino , Equivalencia Terapéutica , Inhibidores del Factor de Necrosis Tumoral/efectos adversos , Inhibidores del Factor de Necrosis Tumoral/farmacocinética
6.
Front Pharmacol ; 11: 609522, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33569002

RESUMEN

Objective: The study aimed to explore the bioequivalence of a proposed biosimilar BAT1806 to its reference products marketed in the EU and US (RoActemra-EU and Actemra-US) among healthy Chinese men. The tolerance, immunogenicity, and pharmacokinetics (PK) of the three drugs were also investigated. Methods: In this randomized, double-blind, single-dose, three-arm, parallel study, a single-dose of 4 mg/kg of the reference products, or the biosimilar was administered to the participants. The participants were followed up for 57 days, and PK, immunogenicity, and tolerance evaluations were completed during this period. Results: The PK parameters were similar in all three groups: BAT1806 (n = 45), RoActemra-EU (n = 42), and Actemra-US (n = 42). The 90% confidence intervals (CIs) for the ratios of C max , AUC0-t and AUC0-∞ were 86.90-104.41% for BAT1806 vs. RoActemra-EU, 91.70-106.15% for BAT1806 vs Actemra-US, and 90.04-105.53% for Actemra-US vs RoActemra-EU. For all comparisons, the 90% CIs for the C max , AUC0-t , and AUC0-∞ were within the predefined bioequivalence limit of 80.00-125.00%. The intersubject variability ranged from 14.5% to 21.5%, which was considerably low. Among the participants, 19 (42.2%), 10 (23.8%), and 12 (28.6%) from the BAT1806, RoActemra-EU, and Actemra-US groups were, respectively, found to be positive for anti-drug antibodies, while 14 (31.1%), nine (21.4%), and 12 (28.6%) were positive for neutralizing antibodies. Nevertheless, these antibodies did not affect the drug concentrations, and the outcomes in the bioequivalence tests were similar after sensitivity analysis. Treatment-related and treatment-emergent adverse events (TEAEs) were recorded in 27, 34, and 32 participants in the BAT1806, RoActemra-EU, and Actemra-US groups, respectively. The most common treatment-related adverse events observed were a decrease in neutrophil, and white blood cell counts. Conclusion: The PK characteristics of BAT1806 were similar to those of the reference products, RoActemra-EU and Actemra-US. Both BAT1806 and the reference products exhibited low intersubject variability and similar safety profiles. Clinical trial registration number: http://www.chinadrugtrials.org.cn/index.html, CTR20180039; https://clinicaltrials.gov/NCT03606876.

7.
Water Sci Technol ; 2017(3): 770-781, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30016295

RESUMEN

A magnetic cation-exchange resin (MCER) was prepared by copolymerization of oleic acid-grafted magnetite with styrene, divinylbenzene (DVB), and triallylisocyanurate (TAIC) for removing Cd(II) from wastewater. A non-magnetic cation-exchange polystyrene resin (CEPR) was also prepared as a reference. Structural and morphological analyses revealed that the MCER and CEPR were mesoporous microspheres; the MCER contained about 25% Fe3O4. The influence of temperature, pH, contact time, and the initial concentration of Cd(II) on the adsorption of Cd(II) was investigated. The maximum adsorption capacity of the MCER reached 88.56 mg/g, which was achieved at 343 K using a Cd(II) initial concentration of 200 mg/L. The adsorption processes attained equilibrium within 120 min for the MCER and 300 min for the CEPR, and were well described by a pseudo-second-order kinetic model. Furthermore, the equilibrium adsorption data fitted the Freundlich isotherm model better than the Langmuir model. The superior magnetic response and regeneration of the MCER make it a good candidate as an adsorbent for removing Cd(II) from wastewater.


Asunto(s)
Cadmio/química , Resinas de Intercambio de Catión/química , Poliestirenos/química , Aguas Residuales/química , Contaminantes Químicos del Agua/química , Adsorción , Cationes/análisis , Óxido Ferrosoférrico , Concentración de Iones de Hidrógeno , Cinética , Temperatura , Purificación del Agua/métodos
8.
Am J Physiol Gastrointest Liver Physiol ; 306(3): G253-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24337009

RESUMEN

Fluorofenidone (AKF-PD) is a novel pyridone agent. The purpose of this study is to investigate the inhibitory effects of AKF-PD on liver fibrosis in rats and the involved molecular mechanism related to hepatic stellate cells (HSCs). Rats treated with dimethylnitrosamine or CCl4 were randomly divided into normal, model, AKF-PD treatment, and pirfenidone (PFD) treatment groups. The isolated primary rat HSCs were treated with AKF-PD and PFD respectively. Cell proliferation and cell cycle distribution were analyzed by bromodeoxyuridine and flow cytometry, respectively. The expression of collagen I and α-smooth muscle actin (α-SMA) were determined by Western blot, immunohistochemical staining, and real-time RT-PCR. The expression of cyclin D1, cyclin E, and p27(kip1) and phosphorylation of MEK, ERK, Akt, and 70-kDa ribosomal S6 kinase (p70S6K) were detected by Western blot. AKF-PD significantly inhibited PDGF-BB-induced HSC proliferation and activation by attenuating the expression of collagen I and α-SMA, causing G0/G1 phase cell cycle arrest, reducing expression of cyclin D1 and cyclin E, and promoting expression of p27(kip1). AKF-PD also downregulated PDGF-BB-induced MEK, ERK, Akt, and p70S6K phosphorylation in HSCs. In rat liver fibrosis, AKF-PD alleviated hepatic fibrosis by decreasing necroinflammatory score and semiquantitative score, and reducing expression of collagen I and α-SMA. AKF-PD attenuated the progression of hepatic fibrosis by suppressing HSCs proliferation and activation via the ERK/MAPK and PI3K/Akt signaling pathways. AKF-PD may be used as a potential novel therapeutic agent against liver fibrosis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/metabolismo , Piridonas/farmacología , Animales , Células Cultivadas , Colágeno/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/patología , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
9.
Antimicrob Agents Chemother ; 57(11): 5344-54, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23959305

RESUMEN

Hepatitis B virus (HBV)-associated chronic liver diseases are treated with nucleoside analogs that target the virus polymerase. While these analogs are potent, drugs are needed to target other virus-encoded gene products to better block the virus replication cycle and chronic liver disease. This work further characterized GLS4 and compared it to the related BAY 41-4109, both of which trigger aberrant HBV core particle assembly, where the virus replication cycle occurs. This was done in HepAD38 cells, which replicate HBV to high levels. In vitro, GLS4 was significantly less toxic for primary human hepatocytes (P < 0.01 up to 100 µM), inhibited virus accumulation in the supernantant of HepAD38 cells (P < 0.02 up to 100 nM), inhibited HBV replicative forms in the liver with a significantly lower 50% effective concentration (EC50) (P < 0.02), and more strongly inhibited core gene expression (P < 0.001 at 100 to 200 nM) compared to BAY 41-4109. In vivo characterization was performed in nude mice inoculated with HepAD38 cells, which grew out as tumors, resulting in viremia. Treatment of mice with GLS4 and BAY 41-4109 showed strong and sustained suppression of virus DNA to about the same extents both during and after treatment. Both drugs reduced the levels of intracellular core antigen in the tumors. Alanine aminotransferase levels were normal. Tumor and total body weights were not affected by treatment. Thus, GLS4 was as potent as the prototype, BAY 41-4109, and was superior to lamivudine, in that there was little virus relapse after the end of treatment and no indication of toxicity.


Asunto(s)
Antivirales/farmacología , ADN Viral/antagonistas & inhibidores , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/tratamiento farmacológico , Pirimidinas/farmacología , Tiazoles/farmacología , Viremia/tratamiento farmacológico , Virión/efectos de los fármacos , Animales , Antivirales/síntesis química , Línea Celular , ADN Viral/biosíntesis , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/ultraestructura , Hepatitis B Crónica/virología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hepatocitos/virología , Humanos , Lamivudine/farmacología , Masculino , Ratones , Ratones Desnudos , Pruebas de Sensibilidad Microbiana , Cultivo Primario de Células , Piridinas/farmacología , Pirimidinas/síntesis química , Tiazoles/síntesis química , Proteínas del Núcleo Viral/antagonistas & inhibidores , Proteínas del Núcleo Viral/metabolismo , Viremia/virología , Virión/ultraestructura , Replicación Viral/efectos de los fármacos
10.
Eur J Clin Invest ; 43(4): 358-68, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23438945

RESUMEN

BACKGROUND: Fluorofenidone (AKF-PD) is a novel pyridone agent. The purpose of this study is to investigate the inhibitory effects of AKF-PD on dimethylnitrosamine (DMN)-induced liver fibrosis in rats and the involved molecular mechanism related to hepatic stellate cells (HSCs). MATERIALS AND METHODS: Wistar rats were randomly divided into normal control, DMN, DMN/AKF-PD treatment and DMN/pirfenidone (PFD) treatment groups. AKF-PD and PFD treatments were, respectively, performed for two activated HSCs lines, rat CFSC-2G and human LX2. The cell proliferation was analysed by MTT. The expression of collagen I was determined by immunohistochemical staining and real-time RT-PCR. The expression of α-smooth muscle actin (α-SMA), tissue inhibitor of metalloproteinases-1 (TIMP-1), extracellular signal regulated kinase (ERK1/2), p38 MAPK (p38), and c-Jun N-terminal kinase/stress-activated protein kinase (JNK) were also detected by real-time RT-PCR and/or Western blot. RESULTS: AKF-PD significantly reduced PDGF-BB-induced proliferation and activation of HSCs, as determined by reducing protein expression of α-SMA and TIMP-1. AKF-PD treatment attenuated PDGF-BB-induced upregulation of phosphorylation of ERK1/2, p38 and JNK. In fibrotic rat liver, AKF-PD reduced the degree of liver injury and hepatic fibrosis, which was associated with reduced the expression of collagen I, α-SMA, TIMP-1 at both mRNA and protein levels. CONCLUSION: AKF-PD treatment inhibits the progression of hepatic fibrosis by suppressing HSCs proliferation and activation via MAPK signalling pathway.


Asunto(s)
Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/prevención & control , Sistema de Señalización de MAP Quinasas/fisiología , Piridonas/farmacología , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Dimetilnitrosamina/toxicidad , Células Estrelladas Hepáticas/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/enzimología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Proteína Quinasa 8 Activada por Mitógenos/genética , Modelos Animales , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Inhibidor Tisular de Metaloproteinasa-1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
11.
Shock ; 38(5): 567-73, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23042199

RESUMEN

Idiopathic pulmonary fibrosis is a progressive, life-threatening, interstitial lung disease with no effective therapy. In this study, we evaluated the effects of fluorofenidone (FD), a novel pyridone agent, on a murine model of bleomycin-induced pulmonary inflammation and fibrosis. Institute for Cancer Research mice were intravenously injected with BLM or saline for 14 consecutive days. Fluorofenidone, pirfenidone (500 mg · kg · d, respectively), or vehicle was administered throughout the course of the experiment. Animals were killed on day 28, and various parameters reflecting pulmonary vascular permeability, influx of inflammatory cells, and levels of transforming growth factor ß in the bronchoalveolar lavage fluid were assessed. Collagen I, α-smooth muscle actin, and fibronectin were measured by real-time reverse transcriptase-polymerase chain reaction or Western blot. Furthermore, caveolin 1 and activation of P38, extracellular signal-regulated kinase, and c-Jun N-terminal kinase were detected by Western blot. Fluorofenidone treatment significantly attenuated the increased pulmonary damage index score, the levels of proteins, transforming growth factor ß, and the influx of cells in bronchoalveolar lavage fluid. Fluorofenidone also markedly reduced the expression of fibronectin, α-smooth muscle actin, and collagen I in mouse lung tissues. Inversely, FD restored caveolin 1 protein and mRNA expression, which was significantly downregulated in BLM-induced lung fibrosis. Fluorofenidone also inhibited phosphorylation of extracellular signal-regulated kinase, P38, and c-Jun N-terminal kinase. These findings collectively suggest that FD is an effective agent with antifibrotic and anti-inflammatory properties, and the mechanisms of its antifibrotic effect include regulating caveolin 1 expression and blocking mitogen-activated protein kinase signaling pathways.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Caveolina 1/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neumonía/metabolismo , Piridonas/farmacología , Actinas/biosíntesis , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Lavado Broncoalveolar , Permeabilidad Capilar/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Fibronectinas/biosíntesis , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Fosforilación/efectos de los fármacos , Neumonía/inducido químicamente , Neumonía/patología , Neumonía/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Pharmazie ; 67(5): 452-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22764581

RESUMEN

Cardiac myofibroblast differentiation, characterized by expression of alpha-smooth muscle actin (alpha-SMA) and fibrillar collagens, plays a key role in the adverse myocardial remodeling. Fluorofenidone (1-(3-fluorophenyl)-5-methyl-2-(1H)-pyridone, AKF-PD) is a novel pyridone antifibrotic agent, which exerts a strong antifibrotic effect. This study investigated the potential role of AKF-PD in suppressing cardiac myofibroblast conversion induced by transforming growth factor-beta1 (TGF-beta1) and the related mitogen-activated protein kinase (MAPK) signaling pathways in neonatal rat cardiac fibroblasts. The MAPK inhibitors used for pathway determination are c-Jun NH(2)-terminal kinase (JNK) inhibitor II (JNK inhibitor), PD98059 (extracellular signal-regulated kinase inhibitor (ERK) inhibitor) and SB203580 (p38 MAPK inhibitor). Cell proliferation was evaluated by multiply-table tournament (MTT) assay. The expressions of fibronectin (FN), alpha-SMA, phosphorylated ERK1/2 (pERK1/2) and ERK1/2 were investigated using Western blot analysis. AKF-PD remarkablely reduced the proliferative response of cardiac fibroblasts by 27.57% compared with TGF-beta1 stimulated group. AKF-PD, PD98059, and JNK inhibitor II completely prevented TGF-beta1-induced FN protein production. In addition, AKF-PD, PD98059 and SB203580 greatly attenuated alpha-SMA expression induced by TGF-beta1. Furthermore, AKF-PD significantly blocked TGF-beta1-induced phosphorylation of ERK. These results indicate that (1) AKF-PD inhibits TGF-beta1-induced myofibroblast differentiation; (2) the anti-fibrotic effects of AKF-PD are partially mediated by ERK phosphorylation.


Asunto(s)
Miofibroblastos/efectos de los fármacos , Piridonas/farmacología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/farmacología , Actinas/biosíntesis , Actinas/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Diferenciación Celular/efectos de los fármacos , Separación Celular , Células Cultivadas , Colágeno/biosíntesis , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibronectinas/biosíntesis , Fibrosis , Flavonoides/farmacología , Indicadores y Reactivos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/citología , Fosforilación , Ratas , Ratas Sprague-Dawley
13.
Antivir Ther ; 17(5): 793-803, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22668794

RESUMEN

BACKGROUND: HBV infection continues to be an important worldwide cause of morbidity and mortality. Patients with chronic hepatitis B can be successfully treated using nucleoside/nucleotide analogues. However, drug-resistant HBV mutants frequently arise, leading to treatment failure and progression to liver disease. Here, we report the effects of GLS4, a non-nucleosidic inhibitor that exhibits a novel and highly specific anti-HBV activity. METHODS: The median inhibitory concentrations (IC(50)s) of GLS4 on HBV were measured by Southern blotting. HBV capsid and core protein levels were detected by immunoblotting. To determine the antiviral activity of GLS4 against adefovir dipivoxil (ADV)-resistant HBV mutants, HepG2 cells transiently transfected with PUC-HBV1.2 plasmids that contained one of three major ADV-resistant mutations (rtA181T, rtA181V and rtN236T) were treated with GLS4. Intracellular HBV replicative intermediates were detected by Southern blotting. The effect on the in vitro assembly of HBV capsid protein was examined using dynamic light scattering and electron microscopy. RESULTS: The IC(50) of GLS4 was 0.012 µM, which is significantly lower than that of lamivudine (0.325 µM). Immunoblot analysis of HepG2.2.15 cells and transiently transfected HepG2 cells indicated that GLS4 treatment interfered with the formation of core particles (assembly). The ADV-resistant HBV mutant strains were also sensitive to GLS4. Upon examining the in vitro assembly of HBV core protein 149 by electron microscopy, increased aberrant particles were observed after GLS4 treatment. CONCLUSIONS: GLS4 is a new and unique potential anti-HBV agent that possesses a different mechanism of action than existing therapeutic drugs.


Asunto(s)
Adenina/análogos & derivados , Antivirales/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/genética , Mutación , Organofosfonatos/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Replicación Viral/efectos de los fármacos , Adenina/farmacología , Antivirales/química , Antivirales/toxicidad , Proteínas de la Cápside/antagonistas & inhibidores , ADN Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Células Hep G2 , Humanos , Concentración 50 Inhibidora , Proteínas del Núcleo Viral/antagonistas & inhibidores , Ensamble de Virus/efectos de los fármacos
14.
Am J Nephrol ; 34(2): 181-94, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21791914

RESUMEN

BACKGROUND: Novel therapeutic agents are urgently needed to combat renal fibrosis. The purpose of this study was to assess, using complete unilateral ureteral obstruction (UUO) in rats, whether fluorofenidone (AKF-PD) [1-(3-fluorophenyl)-5-methyl-2-(1H)-pyridone] inhibits renal fibrosis, and to determine whether it exerts its inhibitory function on renal fibroblast activation. METHODS: Sprague-Dawley rats were randomly divided into 3 groups: sham operation, UUO and UUO/AKF-PD (500 mg/kg/day). Renal function, tubulointerstitium damage index score, extracellular matrix (ECM) deposition, and the expressions of TGF-ß(1), collagen III, α-SMA, p-Smad2, p-Smad3, p-ERK1/2, p-JNK and p-p38 were measured. In addition, the expressions of α-SMA, fibronectin, CTGF, p-Smad2/3, p-ERK1/2, p-p38 and p-JNK were measured in TGF-ß(1)-stimulated normal rat renal fibroblasts (NRK-49F). RESULTS: AKF-PD treatment significantly attenuated tubulointerstitium damage, ECM deposition, the expressions of TGF-ß(1), collagen III, α-SMA, p-ERK1/2, p-p38 and p-JNK in vivo. In vitro, AKF-PD dose-dependently inhibited expressions of α-SMA, fibronectin and CTGF. Furthermore, AKF-PD did not inhibit Smad2/3 phosphorylation or nuclear accumulation, but rather attenuated ERK, p38 and JNK activation. CONCLUSION: AKF-PD treatment inhibits the progression of renal interstitial fibrosis in obstructed kidneys; this is potentially achieved by suppressing fibroblast activation. Therefore, AKF-PD is a special candidate for the treatment of renal fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis/tratamiento farmacológico , Regulación de la Expresión Génica , Túbulos Renales/metabolismo , Piridonas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Peso Corporal , Riñón/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas Smad/metabolismo
15.
Pharmazie ; 66(12): 961-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22312703

RESUMEN

OBJECTIVES: The present study was designed to investigate the potential effects and mechanism of fluorofenidone (AKF-PD) on transforming growth factor beta1 (TGF-beta1)-induced tubular epithelial-mesenchymal transition (EMT) and the expression of connective tissue growth factor (CTGF) in human proximal tubular epithelial cells. METHODS: HK-2 cells were pretreated with AKF-PD, pirfenidone (PFD), Losartan, and SB431542 (an inhibitor of TGF-beta type I receptor). The pretreated HK-2 cells were subsequently co-treated with TGF-beta1 (5 ng/ml). The morphological changes of HK-2 cells were observed under an inverted microscope. Expression of alpha-SMA was detected by Western blot and immunofluorescence. The protein expression of ZO-1, fibronectin, CTGF, phosphorylated Smad2 (p-Smad2) and phosphorylated Smad3 (p-Smad3) were evaluated by Western blot. RESULTS: Through down-regulation of p-Smad2 and p-Smad3 proteins, AKF-PD significantly inhibited protein expression of alpha-SMA, fibronectin, and CTGF. Meanwhile, the depressed ZO-1 expression and morphological changes induced by TGF-beta1 were attenuated by AKF-PD. CONCLUSION: AKF-PD acts as an anti-fibrotic agent through blocking TGF-beta/Smads signaling and consequently inhibits TGF-beta1-induced EMT and CTGF expression in human proximal tubular epithelial cells.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Transición Epitelial-Mesenquimal/efectos de los fármacos , Túbulos Renales Proximales/citología , Piridonas/farmacología , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/fisiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Western Blotting , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Fibronectinas/biosíntesis , Técnica del Anticuerpo Fluorescente , Humanos , Indicadores y Reactivos , Túbulos Renales Proximales/efectos de los fármacos , Losartán/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
16.
Inflammation ; 32(3): 139-45, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19274442

RESUMEN

Tumour necrosis factor alpha (TNF-alpha) is a proinflammatory cytokine, which has been shown to be a causative factor in rheumatoid arthritis, inflammatory bowel disease and septic shock. Proinflammatory effect of TNF-alpha is activated mainly through human TNF receptor-1 (TNF-R1). However, the role of the fourth cystein-rich domain (CRD4) of TNF-R1 extracellular portion in the interaction of TNF-alpha with TNF-R1 is still unclear. In the present study, binding activity of TNF-alpha to TNF-R1 and protein levels of IkappaB-alpha and nuclear transcription factor kappa B (NF-kappaB) p65 subunit in HeLa cells were measured using enzyme-linked immunosorbent assay (ELISA) and western-blot analysis. Pep 3 (LRENECVS) which was derived from the hydrophilic region of A1 module in CRD4 remarkably inhibited the binding of TNF-alpha to TNF-R1, and also reversed TNF-alpha-induced degradation of IkappaB-alpha and nuclear translocation of NF-kappaB p65 subunit in HeLa cells. Our results confirmed that the hydrophilic region of A1 module in CRD4 participated in the interaction of TNF-alpha with TNF-R1, and demonstrated the potential of small-molecule TNF-alpha extracellular inhibitors targeting at A1 module in CRD4 of TNF-R1 in suppressing proinflammatory effect of TNF-alpha.


Asunto(s)
Inflamación/prevención & control , Fragmentos de Péptidos/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/química , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Sitios de Unión , Células HeLa , Humanos , Proteínas I-kappa B/análisis , Inhibidor NF-kappaB alfa , FN-kappa B/análisis , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/uso terapéutico , Unión Proteica/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Neurosci Lett ; 452(2): 90-5, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19154776

RESUMEN

The amyloid precursor protein (APP) is cleaved enzymatically by non-amyloidogenic and amyloidogenic pathways. alpha-Secretase cleaves APP within beta-amyloid protein (Abeta) sequence, resulting in the release of a secreted fragment of APP (sAPPalpha) and precluding Abeta generation. Cryptotanshinone (CTS), an active component of the medicinal herb Salvia miltiorrhiza, has been shown to improve learning and memory in several pharmacological models of Alzheimer's disease (AD). However, the effects of CTS on the Abeta plaque pathology and the APP processing in AD are unclear. Here we reported that CTS strongly attenuated amyloid plaque deposition in the brain of APP/PS1 transgenic mice. In addition, CTS significantly improved spatial learning and memory in APP/PS1 mice assessed by the Morris water maze testing. To define the exact molecular mechanisms involved in the beneficial effects of CTS, we investigated the effects of the CTS on APP processing in rat cortical neuronal cells overexpressing Swedish mutant human APP695. CTS was found to decrease Abeta generation in concentration-dependent (0-10muM) manner. Interestingly, the N-terminal APP cleavage product, sAPPalpha was markedly increased by CTS. Further study showed that alpha-secretase activity was increased by CTS. Taken together, our results suggested CTS improved the cognitive ability in AD transgenic mice and promoted APP metabolism toward the non-amyloidogenic products pathway in rat cortical neuronal cells. CTS shows a promising novel way for the therapy of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide/efectos de los fármacos , Precursor de Proteína beta-Amiloide/efectos de los fármacos , Fenantrenos/farmacología , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Canfanos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Panax notoginseng , Placa Amiloide/efectos de los fármacos , Placa Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Ratas , Salvia miltiorrhiza/química , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
18.
Pharmazie ; 64(11): 755-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20099522

RESUMEN

Angiotensin II (Ang II)-induced hypertrophy response in cultured cardiomyocytes is partially mediated by endothelin-1 (ET-1). Endothelin converting enzyme-1 (ECE-1) is the rate limiting enzyme in the process of ET-1 production. In this study, two peptides which have significant inhibitory effect to the activity of rat ECE-1 purified from stable rat ECE-1-expressed CHO lines, were selected from 13 big ET-1 analogues. We found that treatment of P8 or P9 reversed the increase of hypertrophy genetic markers and cell surface area in primary cultured neonatal rat cardiomyocytes stimulated by Ang II. Besides, depletion of ECE-1 by RNA interference also revealed similar results as P8 or P9 treatment. These results confirmed that ECE-1 plays a key role in regulating Ang II-induced hypertrophy response in cultured cardiomyocytes.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Angiotensina II/toxicidad , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Metaloendopeptidasas/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Ácido Aspártico Endopeptidasas/biosíntesis , Western Blotting , Células CHO , Tamaño de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Endotelina-1/química , Endotelina-1/farmacología , Enzimas Convertidoras de Endotelina , Metaloendopeptidasas/biosíntesis , Miocitos Cardíacos/ultraestructura , Péptidos/aislamiento & purificación , Péptidos/farmacología , Interferencia de ARN/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...