Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biol Toxicol ; 40(1): 38, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789868

RESUMEN

Ensartinib, an approved ALK inhibitor, is used as a first-line therapy for advanced ALK-positive non-small cell lung cancer in China. However, the hepatotoxicity of ensartinib seriously limits its clinical application and the regulatory mechanism is still elusive. Here, through transcriptome analysis we found that transcriptional activation of TXNIP was the main cause of ensartinib-induced liver dysfunction. A high TXNIP level and abnormal TXNIP translocation severely impaired hepatic function via mitochondrial dysfunction and hepatocyte apoptosis, and TXNIP deficiency attenuated hepatocyte apoptosis under ensartinib treatment. The increase in TXNIP induced by ensartinib is related to AKT inhibition and is mediated by MondoA. Through screening potential TXNIP inhibitors, we found that the natural polyphenolic flavonoid rutin, unlike most reported TXNIP inhibitors can inhibit TXNIP by binding to TXNIP and partially promoting its proteasomal degradation. Further studies showed rutin can attenuate the hepatotoxicity of ensartinib without antagonizing its antitumor effects. Accordingly, we suggest that TXNIP is the key cause of ensartinib-induced hepatotoxicity and rutin is a potential clinically safe and feasible therapeutic strategy for TXNIP intervention.


Asunto(s)
Apoptosis , Proteínas Portadoras , Rutina , Animales , Humanos , Masculino , Ratones , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Rutina/farmacología
2.
Autophagy ; 18(5): 1152-1173, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34432562

RESUMEN

Excessive macroautophagy/autophagy is one of the causes of cardiomyocyte death induced by cardiovascular diseases or cancer therapy, yet the underlying mechanism remains unknown. We and other groups previously reported that autophagy might contribute to cardiomyocyte death caused by sunitinib, a tumor angiogenesis inhibitor that is widely used in clinic, which may help to understand the mechanism of autophagy-induced cardiomyocyte death. Here, we found that sunitinib-induced autophagy leads to apoptosis of cardiomyocyte and cardiac dysfunction as the cardiomyocyte-specific Atg7-/+ heterozygous mice are resistant to sunitinib. Sunitinib-induced maladaptive autophagy selectively degrades the cardiomyocyte survival mediator CCN2 (cellular communication network factor 2) through the TOLLIP (toll interacting protein)-mediated endosome-related pathway and cardiomyocyte-specific knockdown of Ccn2 through adeno-associated virus serotype 9 (AAV9) mimics sunitinib-induced cardiac dysfunction in vivo, suggesting that the autophagic degradation of CCN2 is one of the causes of sunitinib-induced cardiotoxicity and death of cardiomyocytes. Remarkably, deletion of Hmgb1 (high mobility group box 1) inhibited sunitinib-induced cardiomyocyte autophagy and apoptosis, and the HMGB1-specific inhibitor glycyrrhizic acid (GA) significantly mitigated sunitinib-induced autophagy, cardiomyocyte death and cardiotoxicity. Our study reveals a novel target protein of autophagic degradation in the regulation of cardiomyocyte death and highlights the pharmacological inhibitor of HMGB1 as an attractive approach for improving the safety of sunitinib-based cancer therapy.


Asunto(s)
Cardiotoxicidad , Factor de Crecimiento del Tejido Conjuntivo , Proteína HMGB1 , Cardiopatías , Sunitinib , Animales , Apoptosis , Autofagia , Cardiotoxicidad/patología , Factor de Crecimiento del Tejido Conjuntivo/genética , Proteína HMGB1/metabolismo , Cardiopatías/inducido químicamente , Cardiopatías/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Sunitinib/farmacología
3.
Expert Opin Drug Metab Toxicol ; 17(11): 1311-1325, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34743659

RESUMEN

INTRODUCTION: By 1 January 2021, the FDA has approved a total of 62 small-molecule kinase inhibitors (SMKIs). The increasing clinical use of small-molecule kinase inhibitors has led to some side effects, the most common of which is cutaneous toxicity, as reflected by approximately 90% (57 of 62) of the FDA-approved SMKIs have reported treatment-related cutaneous toxicities. Since these cutaneous toxicities may have a crucial influence on the emotional, physical and psychosocial health of the patients, it is of great importance for doctors, patients, oncologists and interrelated researchers to be aware of the cutaneous side effects of these drugs in order to make the diagnosis accurate and the treatment appropriate. AREAS COVERED: This review aims to summarize the potential cutaneous toxicities and the frequency of occurrence of FDA-approved 62 SMKIs, and provide a succinct overview of the potential mechanisms of certain cutaneous toxicities. The literature review was performed based on PubMed database and FDA official website. EXPERT OPINION: It is significant to determine the risk factors for SMKI-induced cutaneous toxicity. The mechanisms underlying SMKI-induced cutaneous toxicities remain unclear at present. Future research should focus on the mechanisms of SMKI-induced cutaneous toxicities to find out mechanistically driven therapies.


Asunto(s)
Inhibidores de Proteínas Quinasas , Enfermedades de la Piel , Aprobación de Drogas , Humanos , Inhibidores de Proteínas Quinasas/efectos adversos , Enfermedades de la Piel/inducido químicamente , Estados Unidos , United States Food and Drug Administration
4.
Autophagy ; 17(10): 3221-3237, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33315519

RESUMEN

Liver dysfunction is an outstanding dose-limiting toxicity of gefitinib, an EGFR (epidermal growth factor receptor)-tyrosine kinase inhibitor (TKI), in the treatment of EGFR mutation-positive non-small cell lung cancer (NSCLC). We aimed to elucidate the mechanisms underlying gefitinib-induced hepatotoxicity, and provide potentially effective intervention strategy. We discovered that gefitinib could sequentially activate macroautophagy/autophagy and apoptosis in hepatocytes. The inhibition of autophagy alleviated gefitinib-induced apoptosis, whereas the suppression of apoptosis failed to lessen gefitinib-induced autophagy. Moreover, liver-specific Atg7+/- heterozygous mice showed less severe liver injury than vehicle, suggesting that autophagy is involved in the gefitinib-promoted hepatotoxicity. Mechanistically, gefitinib selectively degrades the important anti-apoptosis factor COX6A1 (cytochrome c oxidase subunit 6A1) in the autophagy-lysosome pathway. The gefitinib-induced COX6A1 reduction impairs mitochondrial respiratory chain complex IV (RCC IV) function, which in turn activates apoptosis, hence causing liver injury. Notably, this autophagy-promoted apoptosis is dependent on PLK1 (polo like kinase 1). Both AAV8-mediated Plk1 knockdown and PLK1 inhibitor BI-2536 could mitigate the gefitinib-induced hepatotoxicity in vivo by abrogating the autophagic degradation of the COX6A1 protein. In addition, PLK1 inhibition could not compromise the anti-cancer activity of gefitinib. In conclusion, our findings reveal the gefitinib-hepatotoxicity pathway, wherein autophagy promotes apoptosis through COX6A1 degradation, and highlight pharmacological inhibition of PLK1 as an attractive therapeutic approach toward improving the safety of gefitinib-based cancer therapy.Abbreviations: 3-MA: 3-methyladenine; AAV8: adeno-associated virus serotype 8; ATG5: autophagy related 5; ATG7: autophagy related 7; B2M: beta-2-microglobulin; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CHX: cycloheximide; COX6A1: cytochrome c oxidase subunit 6A1; c-PARP: cleaved poly(ADP-ribose) polymerase; CQ: chloroquine; GOT1/AST: glutamic-oxaloacetic transaminase 1, soluble; GPT/ALT: glutamic pyruvic transaminase, soluble; HBSS: Hanks´ balanced salt solution; H&E: hematoxylin and eosin; MAP1LC3/LC3: microtubule associated proteins 1 light chain 3; PLK1: polo like kinase 1; RCC IV: respiratory chain complex IV; ROS: reactive oxygen species; TUBB8: tubulin beta 8 class VIII.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Enfermedad Hepática Inducida por Sustancias y Drogas , Neoplasias Pulmonares , Animales , Apoptosis , Autofagia , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas de Ciclo Celular , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Gefitinib/farmacología , Neoplasias Pulmonares/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , Quinasa Tipo Polo 1
5.
Toxicol Lett ; 338: 1-9, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33248157

RESUMEN

Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor indicated for the first-line treatment of patients with metastatic or advanced non-small cell lung cancer (NSCLC) whose tumors have specific EGFR mutations. Pulmonary toxicity is one of the fatal adverse effects of gefitinib and the underlying mechanism remains unclear. Here we demonstrated that alveolar macrophages contributed to gefitinib-induced pulmonary toxicity through promoting alveolar epithelial cells to undergo epithelial to mesenchymal transition (EMT) and inducing activation and antiapoptotic effect in fibroblasts. Further, we found that alveolar macrophage-secreted MCP-1 worked as a key factor in the pathologic changes of these two cell types. Gefitinib increased Mcp-1 transcription level via the nuclear import of the transcription factor STAT3. In conclusion, our data uncovered the underlying mechanisms of macrophage-promoted pulmonary toxicity in the presence of gefitinib. MCP-1 antibody or inhibition of STAT3 activation may represent novel therapeutic strategies for preventing gefitinib-induced pulmonary toxicity.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Antineoplásicos/toxicidad , Comunicación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Gefitinib/toxicidad , Pulmón/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Anciano , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Persona de Mediana Edad , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
6.
Cell Res ; 30(9): 779-793, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32296111

RESUMEN

Hand-foot skin reaction (HFSR), among the most significant adverse effects of sorafenib, has been limiting the clinical benefits of this frontline drug in treating various malignant tumors. The mechanism underlying such toxicity remains poorly understood, hence the absence of effective intervention strategies. In the present study, we show that vascular endothelial cells are the primary cellular target of sorafenib-induced HFSR wherein soluble heparin-binding epidermal growth factor (s-HBEGF) mediates the crosstalk between vascular endothelial cells and keratinocytes. Mechanistically, s-HBEGF released from vascular endothelial cells activates the epidermal growth factor receptor (EGFR) on keratinocytes and promotes the phosphorylation of c-Jun N-terminal kinase 2 (JNK2), which stabilizes sirtuin 1 (SIRT1), an essential keratinization inducer, and ultimately gives rise to HFSR. The administration of s-HBEGF in vivo could sufficiently induce hyper-keratinization without sorafenib treatment. Furthermore, we report that HBEGF neutralization antibody, Sirt1 knockdown, and a classic SIRT1 inhibitor nicotinamide could all significantly reduce the sorafenib-induced HFSR in the mouse model. It is noteworthy that nicotinic acid, a prodrug of nicotinamide, could substantially reverse the sorafenib-induced HFSR in ten patients in a preliminary clinical study. Collectively, our findings reveal the mechanism of vascular endothelial cell-promoted keratinization in keratinocytes and provide a potentially promising therapeutic strategy for the treatment of sorafenib-induced HFSR.


Asunto(s)
Células Endoteliales/metabolismo , Mano/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Queratinocitos/metabolismo , Niacinamida/farmacología , Sirtuina 1/metabolismo , Piel/patología , Sorafenib/efectos adversos , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Pie/patología , Células HaCaT , Humanos , Queratosis/patología , Masculino , Ratones Endogámicos ICR , Persona de Mediana Edad , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos
7.
Toxicol Appl Pharmacol ; 383: 114768, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31639374

RESUMEN

Crizotinib is an oral small-molecule tyrosine kinase inhibitor targeting anaplastic lymphoma kinase (ALK), ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) and MET proto-oncogene, receptor tyrosine kinase (MET). Unfortunately, hepatotoxicity is a serious limitation in its clinical application, and the reason remains largely unknown. In this study, we tested the effect of crizotinib in human hepatocyte cell line HL-7702 and human primary hepatocytes, and the results showed that crizotinib treatment caused hepatocyte damage, suggesting that crizotinib induced liver injury by causing hepatocyte death, consistent with the clinical cases. Mechanistically, crizotinib induced hepatocyte death via the apoptotic pathway, and cleaved PARP (c-PARP) was observed as a signaling protein. Moreover, mitochondrial membrane potential (MMP) decrease contributed to crizotinib-induced hepatocyte apoptosis accompanied by hepatocyte DNA damage and reactive oxygen species (ROS) generation. Importantly, crizotinib induced hepatocyte apoptosis independent of its targets, ALK, ROS1 and MET. In conclusion, our data showed that crizotinib induced liver injury through hepatocyte death via the apoptotic pathway which was independent of ALK, ROS1 and MET. And we also found that MMP decrease, DNA damage and ROS generation were involved in the process.


Asunto(s)
Apoptosis/efectos de los fármacos , Crizotinib/toxicidad , Daño del ADN/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adulto , Apoptosis/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Daño del ADN/fisiología , Relación Dosis-Respuesta a Droga , Hepatocitos/patología , Humanos , Masculino , Inhibidores de Proteínas Quinasas/toxicidad , Proto-Oncogenes Mas
8.
Sensors (Basel) ; 19(4)2019 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-30823553

RESUMEN

Ultra high frequency radio frequency identification (UHF RFID)-based indoor localization technology has been a competitive candidate for context-awareness services. Previous works mainly utilize a simplified Friis transmission equation for simulating/rectifying received signal strength indicator (RSSI) values, in which the directional radiation of tag antenna and reader antenna was not fully considered, leading to unfavorable performance degradation. Moreover, a k-nearest neighbor (kNN) algorithm is widely used in existing systems, whereas the selection of an appropriate k value remains a critical issue. To solve such problems, this paper presents an improved kNN-based indoor localization algorithm for a directional radiation scenario, IKULDAS. Based on the gain features of dipole antenna and patch antenna, a novel RSSI estimation model is first established. By introducing the inclination angle and rotation angle to characterize the antenna postures, the gains of tag antenna and reader antenna referring to direct path and reflection paths are re-expressed. Then, three strategies are proposed and embedded into typical kNN for improving the localization performance. In IKULDAS, the optimal single fixed rotation angle is introduced for filtering a superior measurement and an NJW-based algorithm is advised for extracting nearest-neighbor reference tags. Furthermore, a dynamic mapping mechanism is proposed to accelerate the tracking process. Simulation results show that IKULDAS achieves a higher positioning accuracy and lower time consumption compared to other typical algorithms.

9.
Eur J Pharmacol ; 847: 26-31, 2019 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-30660576

RESUMEN

Renal fibrosis is the common final outcome of nearly all progressive chronic kidney diseases (CKD) that eventually develop into end-stage renal failure, which threatens the lives of patients. Currently, there are no effective drugs for the treatment of renal fibrosis. However, studies have shown that certain plant natural products have a fibrosis-alleviating effect. Thus, we have screened a large number of natural products for their ability to protect against renal fibrosis and found that bisdemethoxycurcumin has a good therapeutic effect in renal fibrosis according to the data obtained in a mouse model of unilateral ureteral obstruction (UUO). The results indicate that bisdemethoxycurcumin can efficiently attenuate renal fibrosis induced by UUO. Additional studies of the bisdemethoxycurcumin mechanism of action in the treatment of renal fibrosis demonstrated that the therapeutic effect of bisdemethoxycurcumin is mediated by the specific induction of fibroblast apoptosis at a concentration of 20 µM. bisdemethoxycurcumin can efficiently protect against renal fibrosis both in vitro and in vivo. This discovery will provide new ideas for renal fibrosis treatment in clinics and a new direction for the development of effective drug therapy of renal fibrosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Curcumina/análogos & derivados , Fibroblastos/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Riñón/efectos de los fármacos , Sustancias Protectoras/farmacología , Insuficiencia Renal Crónica/tratamiento farmacológico , Animales , Productos Biológicos/farmacología , Línea Celular , Curcumina/farmacología , Diarilheptanoides , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Obstrucción Ureteral/tratamiento farmacológico , Sistema Urinario/efectos de los fármacos
10.
Toxicol Appl Pharmacol ; 366: 10-16, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30653976

RESUMEN

Idiopathic pulmonary fibrosis is a pathological result of dysfunctional repair response to tissue injury, leading to chronically impaired gas exchange and death. Macrophages are believed to be critical in this disease pathogenesis; However, the exact mechanisms remain enigmatic. Here, we demonstrated that macrophages might contribute to pulmonary fibrosis at the early stage because the aggregation of macrophages appeared earlier than epithelial-mesenchymal transition and fibrosis in mouse and rat experimental models of pulmonary fibrosis. It has been found that macrophages could promote epithelial-mesenchymal transition of alveolar epithelial cells and fibroblast migration in co-culture models between macrophages and alveolar epithelial cells/fibroblasts. Importantly, we used protein micro array to analyze the cytokines that were altered after bleomycin treatment. Only thymic stromal lymphopoietin and matrix metalloproteinase 9 were significantly increased. We further confirmed that TSLP participated in the macrophage-induced epithelial-mesenchymal transition of alveolar epithelial cells using a TSLP recombinant protein. MMP9 was also involved in macrophage-induced fibroblast migration, which can be reversed by an inhibitor of MMP9. Collectively, these findings explained the underlying mechanisms of macrophage-promoted pulmonary fibrosis.


Asunto(s)
Bleomicina , Citocinas/metabolismo , Fibroblastos/enzimología , Pulmón/enzimología , Macrófagos Alveolares/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Fibrosis Pulmonar/enzimología , Animales , Línea Celular , Movimiento Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Fibroblastos/patología , Pulmón/patología , Macrófagos Alveolares/patología , Ratones Endogámicos ICR , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas Wistar , Transducción de Señal , Factores de Tiempo , Linfopoyetina del Estroma Tímico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA