Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
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.
Arch Toxicol ; 97(3): 635-650, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36773078

RESUMEN

The phosphatidylinositol 3-kinase (PI3K) signalling pathway regulates cell survival, proliferation, migration, metabolism and other vital cellular life processes. In addition, activation of the PI3K signalling pathway is important for cancer development. As a result, a variety of PI3K inhibitors have been clinically developed to treat malignancies. Although several PI3K inhibitors have received approval from the Food and Drug Administration (FDA) for significant antitumour activity, frequent and severe adverse effects have greatly limited their clinical application. These toxicities are mostly on-target and immune-mediated; nevertheless, the underlying mechanisms are still unclear. Current management usually involves intervention through symptomatic treatment, with discontinuation if toxicity persists. Therefore, it is necessary to comprehensively understand these adverse events and ensure the clinical safety application of PI3K inhibitors by establishing the most effective management guidelines, appropriate intermittent dosing regimens and new combination administration. Here, the focus is on the development of PI3K inhibitors in cancer therapy, with particular emphasis on isoform-specific PI3K inhibitors. The most common adverse effects of PI3K inhibitors are also covered, as well as potential mechanisms and management approaches.


Asunto(s)
Antineoplásicos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Neoplasias , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas , Inhibidores de las Quinasa Fosfoinosítidos-3/toxicidad , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Neoplasias/inducido químicamente , Transducción de Señal , Antineoplásicos/farmacología
3.
Bioorg Chem ; 121: 105673, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35217375

RESUMEN

Fibroblast growth factor receptor 4 (FGFR4) together with co-receptors modulate the activation of downstream proteins that regulate fundamental processes, and elevated FGFR4 activity is associated with Hepatocellular Carcinoma (HCC). Hence, FGFR4 is a promising therapeutic target for HCC. Based on BLU9931, we designed and synthesized a series of phenylquinazoline derivatives as novel inhibitors of FGFR4 through the covalent reversible strategy. Among them, a novel compound (C3) showed FGFR4 and cell proliferation inhibitory activity. Cellular mechanism studies demonstrated that compound C3 induced apoptosis via the FGFR4 signaling pathway blockage. Further mechanism study showed that C3 has the reversible covalent binding capacity, could be used as a reference for the development of novel FGFR4 covalent reversible inhibitors.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo
4.
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
5.
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
6.
Clin Exp Pharmacol Physiol ; 45(7): 659-667, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29543385

RESUMEN

With the use of multikinase inhibitors (MKIs) having emerged in recent years, skin toxicities such as hand-foot skin reaction (HFSR) are primary side effects, and they lack effective prediction methods. Here, we updated a previous systematic review by establishing a meta-analysis of the risk of developing HFSR among patients receiving MKIs and antivascular endothelial growth factor antibody. Publications from PubMed and abstracts presented at the American Society of Clinical Oncology Annual Meeting up to February 5, 2015, were searched to identify relevant studies, and a total of 236 patients with metastatic tumours in nine trials were included for analysis. In the meta-analysis, the pooled incidence rates of all-grade and high-grade HFSR among patients who received the combination therapy were 56.9% [95% confidence interval (CI), 45%-71.1%] and 14.3% (95% CI, 9%-24.2%), respectively, with significant differences observed with MKI monotherapy (P < .05). Further subgroup analysis demonstrated that increasing the dosages of bevacizumab (77.8% vs 51.1%, P = .04) and MKIs (64.3% vs 52.6%, P = .02) significantly increased HFSR incidence. Moreover, combination with chemotherapy exerted a minimal effect on HFSR risk (61% vs 55.3%, P = .5). This updated review and meta-analysis confirm the increased risk of HFSR incidence due to the use of MKIs and antivascular endothelial growth factor antibody. Thus, using these therapies requires safety standards.


Asunto(s)
Bevacizumab/farmacología , Pie , Mano , Inhibidores de Proteínas Quinasas/efectos adversos , Piel/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/inmunología , Animales , Bevacizumab/inmunología , Humanos , Riesgo
7.
Br J Cancer ; 116(8): 1027-1036, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28267710

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common types of malignant tumour and has poor prognosis. Currently, systematic chemotherapy is the only approach to prolong survival. Thus the development of new treatment regimens is urgently needed to improve the therapeutic efficacy. Our study intended to assess the combination of dasatinib and irinotecan against HCC and made an effort to develop a potential medical choice for advanced HCC patients. METHODS: We used SRB colorimetric assay and clonogenic assay to assess antitumour effect in vitro and HCC xenograft model to assess antitumour effect in vivo. We applied flow cytometry and western blotting to explore the mechanism of the combined therapy. Knockdown and overexpression of PLK1 are also applied for validation. RESULTS: We confirmed that dasatinib has synergistic effect with irinotecan (or SN38) on HCC both in vitro and in vivo. The effect is due to arisen apoptosis rate of HCC cells that is accompanied by mitochondria dysfunction. The enhanced antitumour efficacy of SN38 could be explained by additional inhibition of PLK1, which is triggered by dasatinib. Unlike existed PLK1 inhibitors, dasatinib does not inhibit PLK1 activity in a direct way. Instead, we found that dasatinib reduces PLK1 level by interfering with its protein synthesis progress. We validated that this kind of downregulation of PLK1 level has a key role in the synergistic effect of the two agents. CONCLUSIONS: Dasatinib is able to reinforce the anti-HCC efficacy of irinotecan/SN38 by downregulation of PLK1 synthesis. The combination of the two agents might be a potential medical choice for HCC therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas de Ciclo Celular/antagonistas & inhibidores , Sinergismo Farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Dasatinib/administración & dosificación , Femenino , Citometría de Flujo , Humanos , Irinotecán , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
8.
BMC Cancer ; 16: 2, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26728137

RESUMEN

BACKGROUND: Chemotherapy is the only therapy option for the majority of AML patients, however, there are several limitations for this treatment. Our aim was to find a new chemotherapy strategy that is more effective and less toxic. METHODS: MTT assays and a xenograft mouse model were employed to evaluate the synergistic activity of all-trans retinoic acid (ATRA) combined with topotecan (TPT). Drug-induced DNA damage and apoptosis were determined by flow cytometry analysis with PI and DAPI staining, the comet assay and Western blots. Short hairpin RNA (shRNA) and a RARα plasmid were used to determine whether RARα expression influenced DNA damage and apoptosis. RESULTS: We found that ATRA exhibited synergistic activity in combination with Topotecan in AML cells, and the enhanced apoptosis induced by Topotecan plus ATRA resulted from caspase pathway activation. Mechanistically, ATRA dramatically down regulated RARα protein levels and led to more DNA damage and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of Topotecan combined with ATRA was further validated in the HL60 xenograft mouse model. CONCLUSIONS: Our data demonstrated, for the first time, that the combination of TPT and ATRA showed potential benefits in AML, providing a novel insight into clinical treatment strategies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Sinergismo Farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Topotecan/administración & dosificación , Tretinoina/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , ARN Interferente Pequeño , Receptores de Ácido Retinoico/biosíntesis , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Molecules ; 22(1)2016 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-28035983

RESUMEN

A series of novel indene-derived retinoic acid receptor α (RARα) agonists have been designed and synthesized. The use of receptor binding, cell proliferation and cell differentiation assays demonstrated that most of these compounds exhibited moderate RARα binding activity and potent antiproliferative activity. In particular, 4-((3-isopropoxy-2,3-dihydro-1H-inden-5-yl)-carbamoyl)benzoic acid (36d), which showed a moderate binding affinity, exhibited a great potential to induce the differentiation of NB4 cells (68.88% at 5 µM). Importantly, our work established indene as a promising skeleton for the development of novel RARα agonists.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Indenos/química , Indenos/síntesis química , Receptor alfa de Ácido Retinoico/agonistas , Línea Celular Tumoral , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Unión Proteica , Relación Estructura-Actividad
10.
Support Care Cancer ; 23(4): 1043-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25277961

RESUMEN

BACKGROUND: This is a single-center, prospective, observational study aiming to determine the effects of unidentified renal insufficiency (URI) on the safety and efficacy of chemotherapy for metastatic colorectal cancer (mCRC) patients. PATIENTS AND METHODS: mCRC patients with normal serum creatinine and who were treated with CapeOx as the first-line therapy were included. Creatinine clearance (CrCL) was estimated using the Cockcroft-Gault formula. URI was characterized by a CrCL of less than 60 ml/min. Logistic regression was used to assess the effects of URI on toxicities and response rates. Kaplan-Meier curve was used to evaluate the effect of URI on survival. RESULTS: A total of 143 patients were enrolled, of whom 34.9 % had URI. Compared with the control group, the URI group had longer toxicity durations and developed significantly more grade 1 to 2 toxicities after adjusting for age, gender, and body mass index. The toxicities include cytopenia (76 vs. 61 %, OR = 1.86, 95 % CI = 0.39-2.53, P < 0.001), diarrhea (34 vs. 29 %, OR = 3.76, 95 % CI = 0.95-6.53, P = 0.007), stomatitis (10 vs. 6 %, OR = 2.81, 95 % CI = 1.10-4.28, P = 0.002), and hand-foot syndrome (18 vs. 11 %, OR = 2.56, 95 % CI = 0.86-5.41, P = 0.045). The response rate and time to progression were significantly lower in the URI group than in the control group (4.5 vs. 5.5 months, HR = 1.57, 95 % CI = 1.09-2.25, P = 0.015), whereas the overall survival rates of the two groups were similar. CONCLUSION: In conclusion, URI can increase the toxicity and decrease the TTP of CapeOx-treated mCRC patients. Renal function screening via CrCL estimation is required for all mCRC patients before initial chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/tratamiento farmacológico , Insuficiencia Renal/etiología , Adulto , Anciano , Neoplasias Colorrectales/mortalidad , Progresión de la Enfermedad , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Seguridad del Paciente , Estudios Prospectivos , Insuficiencia Renal/tratamiento farmacológico , Insuficiencia Renal/mortalidad , Tasa de Supervivencia
11.
Toxicol Appl Pharmacol ; 274(2): 319-27, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24321340

RESUMEN

Combretastatin A-4 (CA-4) has already entered clinical trials of solid tumors over ten years. However, the limited anticancer activity and dose-dependent toxicity restrict its clinical application. Here, we offered convincing evidence that CA-4 induced autophagy in various cancer cells, which was demonstrated by acridine orange staining of intracellular acidic vesicles, the degradation of p62, the conversion of LC3-I to LC3-II and GFP-LC3 punctate fluorescence. Interestingly, CA-4-mediated apoptotic cell death was further potentiated by pretreatment with autophagy inhibitors (3-methyladenine and bafilomycin A1) or small interfering RNAs against the autophagic genes (Atg5 and Beclin 1). The enhanced anticancer activity of CA-4 and 3-MA was further confirmed in the SGC-7901 xenograft tumor model. These findings suggested that CA-4-elicited autophagic response played a protective role that impeded the eventual cell death while autophagy inhibition was expected to improve chemotherapeutic efficacy of CA-4. Meanwhile, CA-4 treatment led to phosphorylation/activation of JNK and JNK-dependent phosphorylation of Bcl-2. Importantly, JNK inhibitor or JNK siRNA inhibited autophagy but promoted CA-4-induced apoptosis, indicating a key requirement of JNK-Bcl-2 pathway in the activation of autophagy by CA-4. We also identified that pretreatment of Bcl-2 inhibitor (ABT-737) could significantly enhance anticancer activity of CA-4 due to inhibition of autophagy. Taken together, our data suggested that the JNK-Bcl-2 pathway was considered as the critical regulator of CA-4-induced protective autophagy and a potential drug target for chemotherapeutic combination.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Sistema de Señalización de MAP Quinasas , Estilbenos/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Compuestos de Bifenilo/farmacología , Línea Celular Tumoral , Humanos , Macrólidos/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Nitrofenoles/farmacología , Fosforilación , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Biomol Ther (Seoul) ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38871446

RESUMEN

Gefitinib is the well-tolerated first-line treatment of non-small cell lung cancer. As it need for analgesics during oncology treatment, particularly in the context ofthe coronavirus disease, where patients are more susceptible to contract high fever and sore throat. This has increased the likelihood of taking both gefitinib and antipyretic analgesic acetaminophen (APAP). Given that gefitinib and APAP overdose can predispose patients to liver injury or even acute liver failure, there is a risk of severe hepatotoxicity when these two drugs are used concomitantly. However, little is known regarding their safety at therapeutic doses. This study simulated the administration of gefitinib and APAP at clinically relevant doses in an animal model and confirmed that gefitinib in combination with APAP exhibited additional hepatotoxicity. We found that gefitinib plus APAP significantly exacerbated cell death, whereas each drug by itself had little or minor effect on hepatocyte survival. Mechanistically, combination of gefitinib and APAP induces hepatocyte death via the apoptotic pathway obviously. Reactive oxygen species (ROS) generation and DNA damage accumulation are involved in hepatocyte apoptosis. Gefitinib plus APAP also promotes the expression of Kelch-like ECH-associated protein 1 (Keap1) and downregulated the antioxidant factor, Nuclear factor erythroid 2-related factor 2 (Nrf2), by inhibiting p62 expression. Taken together, this study revealed the potential ROS-mediated apoptosis-dependent hepatotoxicity effect of the combination of gefitinib and APAP, in which the p62/Keap1/Nrf2 signaling pathway participates and plays an important regulatory role.

13.
Autophagy ; 20(2): 416-436, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37733896

RESUMEN

Crizotinib, a small-molecule tyrosine kinase inhibitor targeting ALK, MET and ROS1, is the first-line drug for ALK-positive metastatic non-small cell lung cancer and is associated with severe, sometimes fatal, cases of cardiac failure, which increases the risk of mortality. However, the underlying mechanism remains unclear, which causes the lack of therapeutic strategy. We established in vitro and in vivo models for crizotinib-induced cardiotoxicity and found that crizotinib caused left ventricular dysfunction, myocardial injury and pathological remodeling in mice and induced cardiomyocyte apoptosis and mitochondrial injury. In addition, we found that crizotinib prevented the degradation of MET protein by interrupting autophagosome-lysosome fusion and silence of MET or re-activating macroautophagy/autophagy flux rescued the cardiomyocytes death and mitochondrial injury caused by crizotinib, suggesting that impaired autophagy activity is the key reason for crizotinib-induced cardiotoxicity. We further confirmed that recovering the phosphorylation of PRKAA/AMPK (Ser485/491) by metformin re-activated autophagy flux in cardiomyocytes and metformin rescued crizotinib-induced cardiomyocyte injury and cardiac complications. In summary, we revealed a novel mechanism for crizotinib-induced cardiotoxicity, wherein the crizotinib-impaired autophagy process causes cardiomyocyte death and cardiac injury by inhibiting the degradation of MET protein, demonstrated a new function of impeded autophagosome-lysosome fusion in drugs-induced cardiotoxicity, pointed out the essential role of the phosphorylation of PRKAA (Ser485/491) in autophagosome-lysosome fusion and confirmed metformin as a potential therapeutic strategy for crizotinib-induced cardiotoxicity.Abbreviations and Acronyms: AAV: adeno-associated virus; ACAC/ACC: acetyl-Co A carboxylase; AMP: adenosine monophosphate; AMPK: AMP-activated protein kinase; ATG5: autophagy related 5; ATG7: autophagy related 7; CHX: cycloheximide; CKMB: creatine kinase myocardial band; CQ: chloroquine; c-PARP: cleaved poly (ADP-ribose) polymerase; DAPI: 4'6-diamidino-2-phenylindole; EF: ejection fraction; FOXO: forkhead box O; FS: fractional shortening; GSEA: gene set enrichment analysis; H&E: hematoxylin and eosin; HF: heart failure; HW: TL: ratio of heart weight to tibia length; IR: ischemia-reperfusion; KEGG: Kyoto encyclopedia of genes and genomes; LAMP2: lysosomal-associated membrane protein 2; LDH: lactate dehydrogenase; MCMs: mouse cardiomyocytes; MMP: mitochondrial membrane potential; mtDNA: mitochondrial DNA; MYH6: myosin, heavy peptide 6, cardiac muscle, alpha; MYH7: myosin, heavy peptide 7, cardiac muscle, beta; NPPA: natriuretic peptide type A; NPPB: natriuretic peptide type B; PI: propidium iodide; PI3K: phosphoinositide 3-kinase; PRKAA/AMPKα: protein kinase AMP-activated catalytic subunit alpha; qPCR: quantitative real-time PCR; SD: standard deviation; SRB: sulforhodamine B; TKI: tyrosine kinase inhibitor; WGA: wheat germ agglutinin.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Metformina , Ratones , Animales , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/genética , Fosforilación , Macroautofagia , Crizotinib/metabolismo , Autofagosomas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cardiotoxicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Péptidos/metabolismo , Miosinas/metabolismo , Lisosomas/metabolismo , Adenosina Monofosfato , Proteínas Tirosina Quinasas Receptoras/metabolismo
14.
Toxicol Lett ; 397: 163-173, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38754640

RESUMEN

Lenvatinib is a multi-target inhibitor that exerts anti-tumor effects by inhibiting angiogenesis and is now commonly used as a first-line treatment for hepatocellular carcinoma. However, with the widespread use of lenvatinib, the problem of serious and fatal hepatotoxicity has become increasingly prominent. Currently, the mechanism behind this toxicity is not yet understood, and as a result, there is a lack of safe and effective intervention strategies with minimal side effects. Here, we established the model of lenvatinib-induced liver injury in vivo and in vitro and found that lenvatinib caused hepatotoxicity by inducing apoptosis. Further mechanistic studies in cellular models revealed that lenvatinib upregulated death receptor signaling pathway, which activated the downstream effector Caspase-8, and ultimately led to apoptosis. Meanwhile, lenvatinib-induced apoptosis was associated with ROS generation and DNA damage. In addition, after screening marketed drugs and natural products in combination with cellular modeling, we identified a potential co-administered drug, dabrafenib, which could alleviate lenvatinib-induced hepatotoxicity. Further mechanistic studies revealed that dabrafenib attenuated lenvatinib-induced hepatotoxicity by inhibiting the activation of the death receptor signaling pathway. Subsequently, cancer cell proliferation assays confirmed that dabrafenib did not antagonize the antitumor effects of lenvatinib. In conclusion, our results validate that apoptosis caused by the death receptor signaling pathway is the key cause of lenvatinib-induced hepatotoxicity, and dabrafenib alleviates lenvatinib-induced hepatotoxicity by inhibiting this pathway.


Asunto(s)
Apoptosis , Enfermedad Hepática Inducida por Sustancias y Drogas , Imidazoles , Oximas , Compuestos de Fenilurea , Quinolinas , Transducción de Señal , Quinolinas/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Oximas/farmacología , Oximas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Humanos , Apoptosis/efectos de los fármacos , Imidazoles/farmacología , Ratones , Masculino , Receptores de Muerte Celular/metabolismo , Antineoplásicos/toxicidad , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Células Hep G2
15.
Mol Carcinog ; 52(1): 18-28, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22006557

RESUMEN

In this paper we report a new myeloid differentiation effect of bortezomib (BTZ) in acute myeloid leukemia (AML) cell lines and primary patient-derived AML cells; this effect was assayed by investigating growth-inhibition, cell morphology, differentiation markers, and nitro-blue tetrazolium reduction. We show that BTZ induces the phosphorylation of several mitogen-activated protein (MAP) kinases, including MEK/ERK, c-Jun N-terminal kinase (JNK), and p38 MAPK. BTZ-induced cell differentiation is almost completely reversed by PD98059, an inhibitor of MEK, which also attenuates the increase in phospho-JNK p46. However, p38 activation does not appear to be required for the differentiation induced by BTZ. Furthermore, the differentiation effect of BTZ is associated with increased protein level of signal transducer and activator of transcription-1 (STAT1), a molecular determinant of myeloid differentiation, due to effects on both its synthesis and degradation. In short, this study reveals that BTZ activates the MEK/ERK cascade, which further up-regulates the expression and activity of the key myeloid transcription factor STAT1, thus promoting myeloid differentiation. These findings contribute to an unexpected potential mechanism for the antitumor activity of BTZ in AML.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Diferenciación Celular/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Pirazinas/farmacología , Factor de Transcripción STAT1/metabolismo , Apoptosis/efectos de los fármacos , Bortezomib , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación , Células Tumorales Cultivadas
16.
Expert Opin Ther Targets ; 27(1): 71-86, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36735300

RESUMEN

INTRODUCTION: Autophagy is a conserved catabolic process that helps recycle intracellular components to maintain homeostasis. The completion of autophagy requires the synergistic effect of multiple canonical autophagic proteins. Defects in autophagy machinery have been reported to promote diseases, rendering autophagy a bone fide health-modifying agent. However, the clinical implication of canonical pan-autophagic activators or inhibitors has often led to undesirable side effects, making it urgent to find a safer autophagy-related therapeutic target. The discovery of non-canonical autophagic proteins has been found to specifically affect the development of diseases without causing a universal impact on autophagy and has shed light on finding a safer way to utilize autophagy in the therapeutic context. AREAS COVERED: This review summarizes recently discovered non-canonical autophagic proteins, how these proteins influence autophagy, and their potential therapeutic role in the disease due to their interaction with autophagy. EXPERT OPINION: Several therapies have been studied thus far and continued research is needed to identify the potential that non-canonical autophagic proteins have for treating certain diseases. In the meantime, continue to uncover new non-canonical autophagic proteins and examine which are likely to have therapeutic implications.


Asunto(s)
Autofagia , Humanos
17.
Toxicol Sci ; 196(2): 152-169, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37702017

RESUMEN

The FDA Modernization Act 2.0 has brought nonclinical drug evaluation into a new era. In vitro models are widely used and play an important role in modern drug development and evaluation, including early candidate drug screening and preclinical drug efficacy and toxicity assessment. Driven by regulatory steering and facilitated by well-defined physiology, novel in vitro skin models are emerging rapidly, becoming the most advanced area in alternative testing research. The revolutionary technologies bring us many in vitro skin models, either laboratory-developed or commercially available, which were all built to emulate the structure of the natural skin to recapitulate the skin's physiological function and particular skin pathology. During the model development, how to achieve balance among complexity, accessibility, capability, and cost-effectiveness remains the core challenge for researchers. This review attempts to introduce the existing in vitro skin models, align them on different dimensions, such as structural complexity, functional maturity, and screening throughput, and provide an update on their current application in various scenarios within the scope of chemical testing and drug development, including testing in genotoxicity, phototoxicity, skin sensitization, corrosion/irritation. Overall, the review will summarize a general strategy for in vitro skin model to enhance future model invention, application, and translation in drug development and evaluation.


Asunto(s)
Dermatitis Fototóxica , Piel , Animales , Evaluación Preclínica de Medicamentos/métodos , Irritantes , Alternativas a las Pruebas en Animales
18.
Biochem Pharmacol ; 208: 115382, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36528067

RESUMEN

Acute respiratory distress syndrome (ARDS) is characterized by noncardiogenic pulmonary edema. It has a high mortality rate and lacks effective pharmacotherapy. With the outbreak of COVID-19 worldwide, the mortality of ARDS has increased correspondingly, which makes it urgent to find effective targets and strategies for the treatment of ARDS. Recent clinical trials of Janus kinase (JAK) inhibitors in treating COVID-19-induced ARDS have shown a positive outcome, which makes the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway a potential therapeutic target for treating ARDS. Here, we review the complex cause of ARDS, the molecular JAK/STAT pathway involved in ARDS pathology, and the progress that has been made in strategies targeting JAK/STAT to treat ARDS. Specifically, JAK/STAT signaling directly participates in the progression of ARDS or colludes with other pathways to aggravate ARDS. We summarize JAK and STAT inhibitors with ARDS treatment benefits, including inhibitors in clinical trials and preclinical studies and natural products, and discuss the side effects of the current JAK inhibitors to reveal future trends in the design of JAK inhibitors, which will help to develop effective treatment strategies for ARDS in the future.


Asunto(s)
COVID-19 , Quinasas Janus , Síndrome de Dificultad Respiratoria , Factores de Transcripción STAT , Humanos , COVID-19/genética , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/genética , Transducción de Señal , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo
19.
Toxicol Lett ; 373: 22-32, 2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-36375637

RESUMEN

Dasatinib, a second-generation BCR-ABL inhibitor, is currently used as first-line treatment for patients with chronic myeloid leukemia. However, dasatinib treatment increases the risk of severe cutaneous toxicity, which limits its long-term safe use in clinic. The underlying mechanism for dasatinib-induced cutaneous toxicity has not been clarified. In this study, we tested the toxicity of dasatinib on human immortal keratinocyte line (HaCaT) and normal human epidermal keratinocytes (NHEK). We found that dasatinib directly caused cytotoxicity on keratinocytes, which could be the explanation of the clinical characteristic of pathology. Mechanistically, dasatinib impaired mitophagy by downregulating HMGB1 protein level in keratinocytes, which led to the accumulation of dysfunctional mitochondria. Mitochondria-derived ROS caused DNA damage and cell apoptosis. More importantly, we confirmed that overexpression of HMGB1 could reverse dasatinib-induced keratinocyte apoptosis, and preliminarily explored the intervention effect of saikosaponin A, which could increase HMGB1 expression, on cutaneous toxicity caused by dasatinib. Collectively, our study revealed that dasatinib induced keratinocyte apoptosis via inhibiting HMGB1-mediated mitophagy and saikosaponin A could be a viable strategy for prevention of dasatinib-induced cutaneous toxicity.


Asunto(s)
Apoptosis , Dasatinib , Humanos , Apoptosis/efectos de los fármacos , Dasatinib/toxicidad , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/efectos de los fármacos , Proteína HMGB1/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Mitofagia/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
20.
Expert Opin Pharmacother ; 24(12): 1361-1373, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37278051

RESUMEN

INTRODUCTION: Alectinib is a second-generation, anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor (TKI) for the treatment of ALK+ non-small cell lung cancer (NSCLC) and is able to induce significant and durable CNS responses. However, long-term use of alectinib has been clinically reported to cause some serious and even life-threatening adverse events. There are currently no effective interventions for its adverse events, and this undoubtedly leads to delays in patient treatment and limits its long-term clinical use. AREAS COVERED: Based on the clinical trials conducted so far, we summarize the efficacy and adverse events that occurred, especially those related to cardiovascular disorders, gastrointestinal disorders, hepatobiliary disorders, musculoskeletal and connective tissue disorders, skin and subcutaneous tissue disorders, and respiratory disorders. The factors that may influence alectinib selection are also described. Findings are based on a PubMed literature search of clinical and basic science research papers spanning 1998-2023. EXPERT OPINION: The significant prolongation of patient survival compared with first-generation ALK inhibitor suggests its potential as a first-line treatment for the NSCLC, but the severe adverse events of alectinib limit its long-term clinical use. Future research should focus on the exact mechanisms of these toxicities, how to alleviate the adverse events caused by alectinib clinically, and the development of next-generation drugs with reduced toxicities.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Quinasa de Linfoma Anaplásico , Carbazoles/efectos adversos , Proteínas Tirosina Quinasas Receptoras/uso terapéutico , Inhibidores de Proteínas Quinasas/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA