Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Acta Pharmacol Sin ; 45(6): 1252-1263, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38360931

ABSTRACT

Although ALK tyrosine kinase inhibitors (ALK-TKIs) have shown remarkable benefits in EML4-ALK positive NSCLC patients compared to conventional chemotherapy, the optimal sequence of ALK-TKIs treatment remains unclear due to the emergence of primary and acquired resistance and the lack of potential prognostic biomarkers. In this study, we systematically explored the validity of sequential ALK inhibitors (alectinib, lorlatinib, crizotinib, ceritinib and brigatinib) for a heavy-treated patient with EML4-ALK fusion via developing an in vitro and in vivo drug testing system based on patient-derived models. Based on the patient-derived models and clinical responses of the patient, we found that crizotinib might inhibit proliferation of EML4-ALK positive tumors resistant to alectinib and lorlatinib. In addition, NSCLC patients harboring the G1269A mutation, which was identified in alectinib, lorlatinib and crizotinib-resistant NSCLC, showed responsiveness to brigatinib and ceritinib. Transcriptomic analysis revealed that brigatinib suppressed the activation of multiple inflammatory signaling pathways, potentially contributing to its anti-tumor activity. Moreover, we constructed a prognostic model based on the expression of IL6, CXCL1, and CXCL5, providing novel perspectives for predicting prognosis in EML4-ALK positive NSCLC patients. In summary, our results delineate clinical responses of sequential ALK-TKIs treatments and provide insights into the mechanisms underlying the superior effects of brigatinib in patients harboring ALKG1269A mutation and resistant towards alectinib, lorlatinib and crizotinib. The molecular signatures model based on the combination of IL6, CXCL1 and CXCL5 has the potential to predict prognosis of EML4-ALK positive NSCLC patients.


Subject(s)
Adenocarcinoma of Lung , Antineoplastic Agents , Lung Neoplasms , Oncogene Proteins, Fusion , Organophosphorus Compounds , Protein Kinase Inhibitors , Pyrimidines , Humans , Organophosphorus Compounds/therapeutic use , Organophosphorus Compounds/pharmacology , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Prognosis , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Lactams/therapeutic use , Carbazoles/therapeutic use , Carbazoles/pharmacology , Sulfones/therapeutic use , Sulfones/pharmacology , Crizotinib/therapeutic use , Crizotinib/pharmacology , Cell Line, Tumor , Piperidines/therapeutic use , Piperidines/pharmacology , Female , Mice , Inflammation/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Male , Anaplastic Lymphoma Kinase/genetics , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/metabolism , Cell Proliferation/drug effects , Mutation , Aminopyridines/therapeutic use , Aminopyridines/pharmacology
2.
Toxics ; 11(11)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37999532

ABSTRACT

Hu7691 represents a novel Pan-Akt kinase inhibitor, demonstrating excellent selectivity towards non-AGC kinase families and pronounced inhibitory effects on the proliferation of multiple tumor cell lines. However, there is currently a notable absence of in vivo toxicological research evidence concerning Hu7691. This study represents the first investigation into the 14-day repeated-dose toxicity of Hu7691 in male and female Sprague Dawley (SD) rats. Male rats were administered daily doses of 12.5, 50, 100, and 150 mg/kg/day, while female rats received doses of 12.5, 25, 50, and 75 mg/kg/day for 14 consecutive days. Hematological assessments, organ weights, and histopathological examinations revealed corresponding alterations, suggesting potential target organs for toxicity including the spleen, thymus, and gastrointestinal tract. It is worth noting that the test substance may also impact the liver, kidneys, heart, and ovaries. The No Observed Effect Level (NOAEL) was determined to be no greater than 12.5 mg/kg/day. Based on the observed gender-related toxicity differences in preliminary trials, it is recommended that the high dose reference dose for male animals in formal experiments should not be less than 100 mg/kg/day, while for female animals, it should be less than 50 mg/kg/day.

3.
Toxics ; 11(2)2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36850989

ABSTRACT

Curcumol, a natural product isolated from the traditional Chinese medicine Rhizoma curcumae, possesses various potential therapeutic values in many diseases. However, evidence of its toxicological profile is currently lacking. In this study, a repeated toxicity study of curcumol was conducted for the first time. SD rats were exposed to doses of 250, 500, 1000 mg/kg in a selected dose formulation for 28 days through oral administration. The potential toxic effects of curcumol on the blood system were observed and further validated in vivo and in vitro. Moreover, other hematology and biochemistry parameters as well as the weight of organs were altered, but no related histopathological signs were observed, indicating these changes were not regarded as toxicologically relevant. Our current findings provide a complete understanding of the safety profile of curcumol, which may contribute to its further study of investigational new drug application.

5.
Phytother Res ; 35(5): 2785-2796, 2021 May.
Article in English | MEDLINE | ID: mdl-33462870

ABSTRACT

Drug-induced liver injury (DILI) has become a significant health care problem worldwide. Centella asiatica (L.) urban was traditionally used to prevent or treat various diseases, yet whether it works on hepatic injury remains unclear. In this study, multiple experimental models with different damage degrees and types of liver injury have been established to evaluate the hepatoprotective effects of an n-butanol extract of Centella asiatica (CA-BU). Our results revealed that CA-BU improved hepatocyte L02 cells survival from H2 O2 -induced oxidative damage in a concentration-dependent manner. We further verified the hepatoprotective effects of CA-BU in mice models of acetaminophen-induced acute liver injury (one of the most common DILIs clinically) and CCl4 -induced acute chemical liver injury, and a rat model of chronic alcoholic steatohepatitis. Furthermore, network pharmacology approaches were performed to explore the underlying mechanisms, and we predicted AKT1, EGFR, VEGFA, and STAT3 as the potential therapeutic targets. In follow-up studies, we will focus on targets verification and provide a deeper insight into the mechanisms of CA-BU against liver damage. Finally, we hope that these findings will provide new ideas and insights for the treatment of acute or chronic liver injury in the clinic.

6.
Mol Oncol ; 14(1): 197-210, 2020 01.
Article in English | MEDLINE | ID: mdl-31721429

ABSTRACT

Hepatocellular carcinoma (HCC) has emerged as one of the most prevalent life-threatening cancers, and the high mortality rate is largely due to the metastasis. The sustained activation of Smad4 and transforming growth factor-ß (TGF-ß) is closely associated with advanced HCC metastasis. However, the regulatory mechanism underlying the aberrant activation of Smad4 and TGF-ß pathway remains elusive. In this study, using a functional screen of USPs siRNA library, we identified ubiquitin-specific proteases USP10 as a deubiquitinating enzyme (DUB) that sustains the protein level of Smad4 and activates TGF-ß signaling. Further analysis showed that USP10 directly interacts with Smad4 and stabilizes it through the cleavage of its proteolytic ubiquitination, thus promoting HCC metastasis. The suppression of USP10 by either shRNAs or catalytic inhibitor Spautin-1 significantly inhibited the migration of HCC cells, whereas the reconstitution of Smad4 was able to efficiently rescue this defect. Overall, our study not only uncovers the regulatory effect of USP10 on the protein abundance of Smad4, but also indicates that USP10 could be regarded as a potential intervention target for the metastatic HCC in Smad4-positive patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination/genetics , Animals , Benzylamines/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Binding , Protein Stability , Quinazolines/pharmacology , RNA, Small Interfering , Smad4 Protein/genetics , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Xenograft Model Antitumor Assays
7.
J Med Chem ; 62(7): 3268-3285, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30856328

ABSTRACT

Multiple sclerosis (MS) is a disease of the autoimmune-mediated disorder in the central nervous system, for which no effective therapeutic agent is currently available. The regulation of macrophage polarization toward M2 is a general benefit for treating MS. The gene biomarker-based phenotypic screening approach was developed, and 3,4-disubstituted piperidine derivative S-28 was identified as a lead compound modulating macrophage M2 polarization. Further SAR studies resulted in the discovery of the most potent modulator D11 that showed good oral bioavailability and significant in vivo therapeutic effects. Mechanistic studies demonstrated that the M2 polarization macrophages modulated by D11 mainly functioned through inhibiting the proliferation of T-cells and activating the phosphorylation of Stat3 and Akt. Therefore, the gene biomarker-based phenotypic screening was demonstrated as a promising tool for the discovery of novel macrophage M2 polarization modulators. Compound D11 may serve as a promising starting point for the development of therapeutics to treat MS.


Subject(s)
Macrophages/drug effects , Multiple Sclerosis/drug therapy , Piperidines/pharmacology , Animals , Biological Availability , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Genetic Markers , High-Throughput Screening Assays , Humans , Mice , Phenotype , Phosphorylation , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship
8.
Br J Cancer ; 117(8): 1192-1201, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28817834

ABSTRACT

BACKGROUND: Metastasis accounts for the most lethal reason for the death of ovarian cancer patients, but remains largely untreated. Epithelial-mesenchymal transition (EMT) is critical for the conversion of early-stage ovarian tumours into metastatic malignancies. Thus the exploration of the signalling pathways promoting EMT would open potential opportunities for the treatment of metastatic ovarian cancer. Herein, the putative role of MDM2 in regulating EMT and metastasis of ovarian cancer SKOV3 cells was investigated. METHODS: The regulatory effects by MDM2 on cell motility was emulated by wound-healing and transwell assays. The effects on EMT transition and Smad pathway were studied by depicting the expression levels of epithelial marker E-cadherin as well as key components of Smad pathway. To evaluate the clinical relevance of our findings, the correlation of MDM2 expression levels with the stages of 104 ovarian cancer patients was investigated by immunohistochemistry assay. RESULTS: We demonstrate that MDM2 functions as a key factor to drive EMT and motility of ovarian SKOV3 cells, by facilitating the activation of TGF-ß-Smad pathway, which results in the increased transcription of snail/slug and the subsequent loss of E-cadherin levels. Such induction of EMT is sustained in either E3 ligase-depleted MDM2 or E3 ligase inhibitor HLI-373-treated cells, while being impaired by the N-terminal deletion of MDM2, which is also reflected by the inhibitory effects against EMT by Nutlin-3a, the N-terminal targeting agent. The expression levels of MDM2 is highly correlated with the stages of the ovarian cancer patients, and the higher expression of MDM2 together with TGFB are closely correlated with poor prognosis and predict a high risk of ovarian cancer patients. CONCLUSIONS: This study suggests that MDM2 activates Smad pathway to promote EMT in ovarian cancer metastasis, and targeting the N-terminal of MDM2 can reprogram EMT and impede the mobility of cancer cells.


Subject(s)
Carcinoma/genetics , Epithelial-Mesenchymal Transition/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Aminoquinolines/pharmacology , Antigens, CD , Blotting, Western , Cadherins/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/drug effects , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunohistochemistry , Neoplasm Metastasis , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , Smad Proteins/metabolism , Snail Family Transcription Factors/genetics , Thymine/analogs & derivatives , Thymine/pharmacology , Transforming Growth Factor beta/metabolism
9.
Int Immunopharmacol ; 36: 256-262, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27179305

ABSTRACT

Interleukin-10 (IL-10)-deficient mice spontaneously develop T cell-mediated colitis. Previous reports have shown that Matrine may reduce the symptoms of acute colitis induced by trinitrobenzene sulfonic acid (TNBS). However, whether Matrine impacts chronic colitis remains unknown. In this study, we investigated whether Matrine could limit the symptoms of spontaneously developed colitis and its potential molecular mechanisms. IL-10 deficient mice were given Matrine or a PBS control by oral gavage daily for 4weeks and were euthanized at week 2 or week 4. We measured body weight, colon length and weight, and histological scores. We also evaluated the spontaneous secretion of IL-12/23p40, IFN-γ and IL-17 in colon explant cultures as well as IFN-γ and IL-17 secretion in unseparated mesenteric lymph node (MLN) cells, and assessed IFN-γ, IL-17, IL-1ß and IL-6 mRNA expression in colon tissue. In addition, we analyzed the proportions of CD4-positive and CD8-positive cells in unseparated MLN cells. Our results show that Matrine-treated mice exhibited better body weight recovery than controls and that histological scores and spontaneously secreted IL-12/23p40, IFN-γ and IL-17 in colon tissue were significantly decreased in treated mice compared with controls. The proportion of CD4-positive cells of MLNs in treated mice was significantly smaller than that in controls at week 4. Both cytokine production and mRNA expression of IFN-γ and IL-17 were significantly reduced in treated mice compared with controls. Taken together, our results indicate that Matrine may ameliorate spontaneously developed chronic colitis and could be considered as a therapeutic alternative for chronic colitis.


Subject(s)
Alkaloids/therapeutic use , Anti-Inflammatory Agents/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Colitis/drug therapy , Quinolizines/therapeutic use , Sophora/immunology , Administration, Oral , Animals , Body Weight/drug effects , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Down-Regulation , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Matrines
10.
Oncotarget ; 7(6): 6933-47, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26771844

ABSTRACT

Although hypoxia is a prominent feature contributing to the therapeutic resistance of hepatocellular carcinoma cells (HCC) against chemotherapeutic agents, including the Topoisomerase I inhibitor SN38, the underlying mechanism is not fully understood and its understanding remains a major clinical challenge. In the present study, we found that hypoxia-induced nuclear translocation and accumulation of YAP acted as a survival input to promote resistance to SN38 in HCC. The induction of YAP by hypoxia was not mediated by HIF-1α because manipulating the abundance of HIF-1α with CoCl2, exogenous expression, and RNA interference had no effect on the phosphorylation or total levels of YAP. The mevalonate-HMG-CoA reductase (HMGCR) pathway may modulate the YAP activation under hypoxia. Combined YAP inhibition using either siRNA or the HMGCR inhibitor statins together with SN38 treatment produced improved anti-cancer effects in HCC cells. The increased anti-cancer effect of the combined treatment with statins and irinotecan (the prodrug of SN-38) was further validated in a human HepG2 xenograft model of HCC in nude mice. Taken together, our findings identify YAP as a novel mediator of hypoxic-resistance to SN38. These results suggest that the administration of SN28 together with the suppression of YAP using statins is a promising strategy for enhancing the treatment response in HCC patients, particularly in advanced stage HCC cases presenting hypoxic resistance.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Camptothecin/analogs & derivatives , Carcinoma, Hepatocellular/pathology , Cell Nucleus/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Hypoxia/physiopathology , Liver Neoplasms/pathology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Blotting, Western , Camptothecin/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Fluorescent Antibody Technique , Humans , Hypoxia/complications , Immunoenzyme Techniques , Irinotecan , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Nude , Phosphoproteins/genetics , Protein Transport , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
11.
Int J Cancer ; 135(7): 1721-32, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24615207

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA) is a promising histone deacetylase (HDAC) inhibitor approved by the US Food and Drug Administration (FDA) and whose clinical application for solid tumours is partially limited by decreased susceptibility in cancer cells due to nuclear factor (NF)-κB activation. As an NF-κB inhibitor, celastrol exhibits potent anticancer effects but has failed to enter clinical trials due to its toxicity. In this report, we demonstrated that the combination of celastrol and SAHA exerted substantial synergistic efficacy against human cancer cells in vitro and in vivo accompanied by enhanced caspase-mediated apoptosis. This drug combination inhibited the activation of NF-κB caused by SAHA monotherapy and consequently led to increased apoptosis in cancer cells. Interestingly, E-cadherin was dramatically downregulated in celastrol-resistant cancer cells, and E-cadherin expression was closely related to decreased sensitivity to celastrol. However, our combination treatment significantly augmented the expression of E-cadherin, suggesting that mutual mechanisms contributed to the synergistic anticancer activity. Furthermore, the enhanced anticancer efficacy of celastrol combined with SAHA was validated in a human lung cancer 95-D xenograft model without increased toxicity. Taken together, our data demonstrated the synergistic anticancer effects of celastrol and SAHA due to their reciprocal sensitisation, which was simultaneously regulated by NF-κB and E-cadherin; thus, the combination of celastrol and SAHA was superior to other combination regimens that rely on a single mechanism. Our findings not only open new opportunities for the clinical development of SAHA but should also motivate the clinical investigation of celastrol, which has been hampered by its toxicity.


Subject(s)
Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Lung Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cadherins/genetics , Cell Proliferation/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , In Vitro Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Pentacyclic Triterpenes , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tripterygium/chemistry , Tumor Cells, Cultured , Vorinostat
12.
Eur J Pharmacol ; 729: 45-53, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24561043

ABSTRACT

Epithelial-mesenchymal transition (EMT) is regarded as the most important mechanism behind the initiation of cancer metastasis. Though there has been great interest in developing therapies aimed at impairing the process of EMT, only few molecules have been identified to orchestrate it so far. Here we report that the dual PI3K/mTOR inhibitor NVP-BEZ235 is capable of preventing human ovarian cancer cell line SKOV-3 and prostatic cancer cell line PC-3 from hypoxia- and TGF-ß1-induced EMT. The addition of NVP-BEZ235 reverses the EMT-like morphologic changes, down-regulation of E-cadherin, and enhancement of cell migration induced by 1% O2 partially through interfering with the expression and transcriptional activity of Hif-1α via PI3K/mTOR pathway. In addition, NVP-BEZ235 inhibits TGF-ß1-induced phosphorylation of Smad2/3 and Akt/GSK-3ß, reduces the expression of Snail both in transcriptional and post-translational level, and consequently prevents the repression of E-cadherin expression as well as the increase of cell motility caused by TGF-ß1. Moreover, in nude mice bearing SKOV-3 ovarian cancer xenografts, NVP-BEZ235 significantly increases the mRNA level of E-cadherin. Taken together, our study demonstrates, for the first time, NVP-BEZ235 can prevent microenvironment and growth factor induced EMT, which suggests this agent as a potential candidate for cancer metastasis treatment.


Subject(s)
Cell Movement/physiology , Epithelial-Mesenchymal Transition/physiology , Imidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Quinolines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transforming Growth Factor beta1/toxicity , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Humans , Mice , Mice, Nude , Phosphatidylinositol 3-Kinase/metabolism , Random Allocation , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays/methods
13.
Food Chem Toxicol ; 51: 251-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22989704

ABSTRACT

Nickel, a metal commonly found in battery plants and welding factories, has potential cardiotoxicity, while all-trans retinoid acid (atRA) can promote cardiovascular repair and myocardial recovery. The purpose of this study was to investigate whether atRA could prevent cardiotoxicity induced by nickel both in vitro and in vivo. In the study, a rat myocardial cell line (H9c2) exposed to different concentrations of nickel chloride (NiCl(2)) displayed apoptotic features accompanied by reactive oxygen species generation. In addition, NiCl(2) also caused obvious apoptosis and systolic dysfunction in primary myocardial cells. Treatment with atRA efficiently attenuated the cytotoxicities triggered by NiCl(2) as it significantly mitigated ROS generation and decreased MAP kinases activity in NiCl(2)-treated cardiomyocytes. Additionally, NiCl(2) exposure caused obvious arrhythmia in Sprague-Dawley rats with the maximum tolerance dose of NiCl(2) between 2 and 3mg/kg. A combinational intragastric administration of 40mg/kg atRA can partially reverse NiCl(2)-induced arrhythmia in rats. Our results suggested that atRA might have therapeutic potential in alleviating the adverse effects of nickel on the cardiovascular system.


Subject(s)
Myocytes, Cardiac/drug effects , Nickel/toxicity , Protective Agents/pharmacology , Tretinoin/pharmacology , Animals , Apoptosis/drug effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/prevention & control , Cell Line , Dose-Response Relationship, Drug , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
14.
Toxicol Appl Pharmacol ; 261(3): 280-91, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22538170

ABSTRACT

Dasatinib, a multitargeted inhibitor of BCR-ABL and SRC kinases, exhibits antitumor activity and extends the survival of patients with chronic myeloid leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL). However, some patients suffer from hepatotoxicity, which occurs through an unknown mechanism. In the present study, we found that Dasatinib could induce hepatotoxicity both in vitro and in vivo. Dasatinib reduced the cell viability of rat primary hepatocytes, induced the release of alanine aminotransferase (ALT) and lactate dehydrogenase (LDH) in vitro, and triggered the ballooning degeneration of hepatocytes in Sprague-Dawley rats in vivo. Apoptotic markers (chromatin condensation, cleaved caspase-3 and cleaved PARP) were detected to indicate that the injury induced by Dasatinib in hepatocytes in vitro was mediated by apoptosis. This result was further validated in vivo using terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assays. Here we found that Dasatinib dramatically increased the level of reactive oxygen species (ROS) in hepatocytes, reduced the intracellular glutathione (GSH) content, attenuated the activity of superoxide dismutase (SOD), generated malondialdehyde (MDA), a product of lipid peroxidation, decreased the mitochondrial membrane potential, and activated nuclear factor erythroid 2-related factor 2 (Nrf2) and mitogen-activated protein kinases (MAPK) related to oxidative stress and survival. These results confirm that oxidative stress plays a pivotal role in Dasatinib-mediated hepatotoxicity. N-acetylcysteine (NAC), a typical antioxidant, can scavenge free radicals, attenuate oxidative stress, and protect hepatocytes against Dasatinib-induced injury. Thus, relieving oxidative stress is a viable strategy for reducing Dasatinib-induced hepatotoxicity.


Subject(s)
Apoptosis/drug effects , Hepatocytes/drug effects , Oxidative Stress/drug effects , Protein Kinase Inhibitors/toxicity , Pyrimidines/toxicity , Thiazoles/toxicity , Acetylcysteine/pharmacology , Alanine Transaminase/blood , Animals , Antioxidants/pharmacology , Blotting, Western , Cell Separation , DNA/chemistry , Dasatinib , Electrophoresis, Gel, Pulsed-Field , Fluorescent Dyes , In Situ Nick-End Labeling , Indoles , L-Lactate Dehydrogenase/blood , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Swelling/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...