Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cardiovasc Res ; 119(10): 1997-2013, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37267414

ABSTRACT

AIMS: Novel cancer therapies leading to increased survivorship of cancer patients have been negated by a concomitant rise in cancer therapies-related cardiovascular toxicities. Sunitinib, a first line multi-receptor tyrosine kinase inhibitor, has been reported to cause vascular dysfunction although the initiating mechanisms contributing to this side effect remain unknown. Long non-coding RNAs (lncRNAs) are emerging regulators of biological processes in endothelial cells (ECs); however, their roles in cancer therapies-related vascular toxicities remain underexplored. METHODS AND RESULTS: We performed lncRNA expression profiling to identify potential lncRNAs that are dysregulated in human-induced pluripotent stem cell-derived ECs (iPSC-ECs) treated with sunitinib. We show that the lncRNA hyaluronan synthase 2 antisense 1 (HAS2-AS1) is significantly diminished in sunitinib-treated iPSC-ECs. Sunitinib was found to down-regulate HAS2-AS1 by an epigenetic mechanism involving hypermethylation. Depletion of HAS2-AS1 recapitulated sunitinib-induced detrimental effects on iPSC-ECs, whereas CRISPR-mediated activation of HAS2-AS1 reversed sunitinib-induced dysfunction. We confirmed that HAS2-AS1 stabilizes the expression of its sense gene HAS2 via an RNA/mRNA heteroduplex formation. Knockdown of HAS2-AS1 led to reduced synthesis of hyaluronic acid (HA) and up-regulation of ADAMTS5, an enzyme involved in extracellular matrix degradation, resulting in disruption of the endothelial glycocalyx which is critical for ECs. In vivo, sunitinib-treated mice showed reduced coronary flow reserve, accompanied by a reduction in Has2os and degradation of the endothelial glycocalyx. Finally, we identified that treatment with high molecular-weight HA can prevent the deleterious effects of sunitinib both in vitro and in vivo by preserving the endothelial glycocalyx. CONCLUSIONS: Our findings highlight the importance of lncRNA-mediated regulation of the endothelial glycocalyx as an important determinant of sunitinib-induced vascular toxicity and reveal potential novel therapeutic avenues to attenuate sunitinib-induced vascular dysfunction.


Subject(s)
RNA, Long Noncoding , Humans , Animals , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Glycocalyx/metabolism , Endothelial Cells/metabolism , Sunitinib/toxicity , Sunitinib/metabolism
2.
Chemosphere ; 308(Pt 2): 136354, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36087734

ABSTRACT

Recently, the potential toxic effects of various pharmaceuticals on the thyroid endocrine system have raised considerable concerns. In this study, we evaluated the adverse effects of sorafenib and sunitinib, two widely used anti-tumor drugs, on the developmental toxicities and thyroid endocrine disruption by using zebrafish (Danio rerio) model. Zebrafish embryos/larvae were exposed to different contentions (0, 10, 50 and 100 nM) of sorafenib and sunitinib for 96 hpf. The results revealed that waterborne exposure to sorafenib and sunitinib exhibited remarkable toxic effects on the survival and development in zebrafish embryos/larvae, which was accompanied by obvious disturbances of thyroid endocrine system (e.g., decreased T3 and T4 content, increased TSH content) and genes' transcription changes within the hypothalamus-pituitary-thyroid (HPT) axis. In addition, we verified a strikingly abnormal thyroid gland organogenesis in zebrafish larvae in response to sorafenib and sunitinib, by assessing the development of thyroid follicles using the WISH staining of tg, the Tg (tg:GFP) zebrafish transgenic line, and histopathological analysis. Taken together, our results indicated sorafenib and sunitinib exposure could induce obvious developmental toxicities and thyroid function disruption in zebrafish embryos/larvae, which might involve a regulatory mechanism, at least in part, by destroying the thyroid follicle structure, and by disturbing the balance of the HPT axis.


Subject(s)
Endocrine Disruptors , Water Pollutants, Chemical , Animals , Endocrine Disruptors/toxicity , Larva , Pharmaceutical Preparations , Protein Kinase Inhibitors/pharmacology , Sorafenib/toxicity , Sunitinib/toxicity , Thyroid Gland , Thyrotropin , Water Pollutants, Chemical/toxicity , Zebrafish/physiology
3.
J Cardiovasc Pharmacol ; 79(6): 799-807, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35266920

ABSTRACT

ABSTRACT: Sunitinib is associated with cardiotoxicity through inhibition of AMP-protein kinase (AMPK) signaling. By contrast, the common antidiabetic agent metformin has demonstrated cardioprotection through indirect AMPK activation. In this study, we investigate the effects of metformin during sunitinib-induced cytotoxicity. Left ventricular developed pressure, coronary flow, heart rate, and infarct size were measured in Langendorff-perfused rat hearts treated with 1 µM sunitinib ±50 µM metformin ±1 µM human equilibrative nucleoside transporter inhibitor S-(4-Nitrobenzyl)-6-thioinosine (NBTI). Western blot analysis was performed for p-AMPKα levels. Primary isolated cardiac myocytes from the left ventricular tissue were used to measure live cell population levels. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to assess adjunctive treatment of and metformin in human hepatoma G2 and promyelocytic leukemia (HL-60) cells treated with 0.1-100 µM sunitinib ±50 µM metformin. In the perfused hearts, coadministration of metformin attenuated the sunitinib-induced changes to left ventricular developed pressure, infarct size, and cardiac myocyte population. Western blot analysis revealed a significant decrease in p-AMPKα during sunitinib treatment, which was attenuated after coadministration with metformin. All metformin-induced effects were attenuated, and NBTI was coadministered. The MTT assay demonstrated an increase in the EC50 value during coadministration of metformin with sunitinib compared with sunitinib monotherapy in hepatoma G2 and HL-60 cell lines, demonstrating the impact and complexity of metformin coadministration and the possible role of AMPK signaling. This study highlights the novel cardioprotective properties of metformin and AMPK activation during sunitinib-induced cardiotoxicity when administered together in the Langendorff heart model.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Metformin , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/metabolism , Adenylate Kinase/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Cardiotoxicity , Infarction/metabolism , Liver Neoplasms/metabolism , Metformin/pharmacology , Myocytes, Cardiac , Rats , Sunitinib/metabolism , Sunitinib/toxicity
4.
Theranostics ; 11(8): 3830-3838, 2021.
Article in English | MEDLINE | ID: mdl-33664864

ABSTRACT

Anti-angiogenics drugs in clinical use for cancer treatment induce cardiotoxic side effects. The endothelin axis is involved in hypertension and cardiac remodelling, and addition of an endothelin receptor antagonist to the anti-angiogenic sunitinib was shown to reduce cardiotoxicity of sunitinib in mice. Here, we explored further the antidote effect of the endothelin receptor antagonist macitentan in sunitinib-treated animals on cardiac remodeling. Methods: Tumor-bearing mice treated per os daily by sunitinib or vehicle were imaged before and after 1, 3 and 6 weeks of treatment by positron emission tomography using [18F]fluorodeoxyglucose and by echocardiography. Non-tumor-bearing animals were randomly assigned to be treated per os daily by vehicle or sunitinib or macitentan or sunitinib+macitentan, and imaged by echocardiography after 5 weeks. Hearts were harvested for histology and molecular analysis at the end of in vivo exploration. Results: Sunitinib treatment increases left ventricular mass and ejection fraction and induces cardiac fibrosis. Sunitinib also induces an early increase in cardiac uptake of [18F]fluorodeoxyglucose, which is significantly correlated with increased left ventricular mass at the end of treatment. Co-administration of macitentan prevents sunitinib-induced hypertension, increase in ejection fraction and cardiac fibrosis, but fails to prevent increase of the left ventricular mass. Conclusion: Early metabolic changes predict sunitinib-induced cardiac remodeling. Endothelin blockade can prevent some but not all cardiotoxic side-effects of sunitinib, in particular left ventricle hypertrophy that appears to be induced by sunitinib through an endothelin-independent mechanism.


Subject(s)
Cardiomegaly/chemically induced , Endothelins/physiology , Sunitinib/toxicity , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Disease Models, Animal , Endothelin Receptor Antagonists/administration & dosage , Female , Fibrosis , Glycolysis/drug effects , Hypertension/chemically induced , Hypertension/prevention & control , Mice , Mice, Inbred C57BL , Mice, Nude , Precision Medicine , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
5.
Hum Exp Toxicol ; 39(12): 1737-1746, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32677474

ABSTRACT

Cutaneous side effects associated with sunitinib use are a major problem in patients receiving cancer treatment. The aim of this study was to investigate the protective effect of adenosine triphosphate (ATP) against possible skin damage resulting from sunitinib use in rats. Thirty Albino Winstar rats were divided into the following three groups: healthy controls (HCs, n = 10), sunitinib (SUN, n = 10), and sunitinib + ATP (SAT, n = 10). ATP was injected intraperitoneally at a dose of 2 mg/kg. One hour subsequent to the administration of ATP and 0.9% NaCl, the SAT and SUN groups were orally administered a dose of 25 mg/kg sunitinib to the stomach. Macroscopic evaluation of the skin indicated lower levels of skin damage in the SAT group than in the SUN group. As an indicator of oxidative stress, malondialdehyde (MDA), total oxidant status (TOS), and oxidative stress index (OSI) levels were significantly higher in the SUN group than in the HC group, while total glutathione (tGSH) and total antioxidant status (TAS) levels were significantly lower. However, MDA, TOS, and OSI levels were significantly lower in the SAT group than in the SUN group, while tGSH and TAS levels were significantly higher. Histopathological examination revealed keratin plugs with edema, vasopathology, and inflammatory cell infiltration in the SUN group. The SAT group showed less necrotic epithelium, keratin plugs, edema, and vasopathology than the SUN group. ATP can be effective in preventing skin damage caused by sunitinib use by reducing oxidative stress.


Subject(s)
Adenosine Triphosphate/therapeutic use , Antineoplastic Agents/toxicity , Protective Agents/therapeutic use , Skin Diseases/chemically induced , Skin Diseases/drug therapy , Skin/drug effects , Sunitinib/toxicity , Adenosine Triphosphate/pharmacology , Animals , Glutathione/metabolism , Male , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Rats, Wistar , Skin/injuries , Skin/metabolism , Skin/pathology , Skin Diseases/metabolism , Skin Diseases/pathology
6.
Life Sci ; 257: 118072, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32659367

ABSTRACT

AIMS: Sunitinib (Su), a tyrosine kinase inhibitor, is one of the most commonly used anti-angiogenic drugs. Some studies have described retinal detachment and photoreceptor damage following systemic exposure to Su, despite beneficial effects achieved with local treatment of ocular pathologies. The aim of this study was to explore the role of NADPH oxidase system and oxidative stress in eyes from Su-treated animals. MAIN METHODS: Male Wistar rats were administered 25 mg Su/kg body weight/day incorporated in the chow for 3 weeks. Upon treatment completion, NADPH oxidase activity and ROS levels were measured in ocular tissue by chemiluminescence and dihydroethidium (DHE) staining, respectively. The expression of NADPH oxidase isoforms (NOX1, NOX2 and NOX4), antioxidant enzymes and endothelial/inducible nitric oxidase isoforms (eNOS/iNOS) in the eyecup and/or retina were measured via immunofluorescence, immunoblotting and RT-qPCR. KEY FINDINGS: NADPH oxidase activity/expression increased in eyecup and retinas from Su-treated rats. Immunohistofluorescence studies in retinal layer confirmed a higher signal of NADPH oxidase isoforms after Su treatment. Treated animals also presented with reductions in NO levels and eNOS expression, whereas iNOS was upregulated. Finally, a significant depletion of antioxidant enzyme glutathione peroxidase was measured in eyecups of rats following Su exposure, and the opposite pattern was seen for glutathione reductase and superoxide dismutase. SIGNIFICANCE: This study demonstrates that Su treatment is associated with NADPH oxidase-derived oxidative stress in the eye. Long-term treatment of Su should be properly monitored to avoid retinotoxic effects that might result in ocular pathologies and sight-threatening conditions.


Subject(s)
Oxidative Stress/drug effects , Protein Kinase Inhibitors/toxicity , Retina/drug effects , Sunitinib/toxicity , Animals , Antioxidants/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Male , NADPH Oxidases/metabolism , Rats , Rats, Wistar , Retina/pathology , Superoxide Dismutase/metabolism
7.
Hum Exp Toxicol ; 39(8): 1046-1053, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32131635

ABSTRACT

In this study, we aimed to show the effect of adenosine 5'-triphosphate (ATP) on sunitinib-induced cardiac injury in rats. The rats (n = 30) were divided equally into three groups as sunitinib group (SG), sunitinib plus ATP group (SAG), and healthy group (HG); 2 mg/kg ATP was injected intraperitoneally (ip) to the SAG group. Same volume normal saline as solvent was administered ip to the other two groups. After 1 h, 25 mg/kg sunitinib was applied orally via catheter to stomach in the SAG and SG groups. This procedure was repeated once daily for 5 weeks. At the end of this period, all animals were sacrificed and their cardiac tissue was removed. Malondialdehyde (MDA), total glutathione (tGSH), tumor necrosis factor α (TNF-α), and nuclear factor κB (NF-κB) levels in rats' cardiac tissues and troponin I (Tp-I) levels in rats' blood samples were evaluated. Histopathological analysis was also performed in cardiac tissues of the animals. MDA, TNF-α, NF-κB, and Tp-I levels were higher in the SG group compared to the SAG and HG groups (p < 0.001). tGSH levels of the SG group were lower than the SAG and HG groups (p < 0.001). The structure and morphology of cardiac muscle fibers and blood vessels were normal in the control group. In the SG group, obvious cardiac muscle tissue damage with dilated myofibers, locally atrophic myofibers, and congested blood vessels were observed. In the SAG group, marked amelioration in these findings was observed. We showed this for the first time that ATP administration exerts a protective effect against cardiac effects of sunitinib.


Subject(s)
Adenosine Triphosphate/therapeutic use , Antineoplastic Agents/toxicity , Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Cardiotoxins/toxicity , Protein Kinase Inhibitors/toxicity , Sunitinib/toxicity , Adenosine Triphosphate/pharmacology , Animals , Cardiotonic Agents/pharmacology , Cardiotoxicity/blood , Cardiotoxicity/metabolism , Glutathione/metabolism , Male , Malondialdehyde/metabolism , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/metabolism , Rats, Wistar , Troponin I/blood , Tumor Necrosis Factor-alpha/metabolism
8.
Eur J Clin Pharmacol ; 76(4): 579-587, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31932871

ABSTRACT

PURPOSE: Sunitinib and pazopanib, two tyrosine kinase inhibitors (TKI), may be targets of potential pharmacokinetic drug-drug interactions (P-PK-DDIs). While strong cytochrome P4503A (CYP3A4) inhibitors or inducers should cause a clinically relevant modification in plasma TKI concentrations, the effect of weak inhibitors is unknown. The objective of this study was to evaluate the association between weak P-PK-DDI and clinically relevant toxicity in real life. PATIENTS AND METHODS: This was a single-center retrospective study including patients treated with sunitinib or pazopanib for any malignancies, for whom a PK-DDI analysis was performed before starting TKI. The primary endpoint was the correlation between P-PK-DDIs and a dose decrease after 1 month of treatment. The secondary endpoint was the correlation between PK-DDIs and drug withdrawal due to toxicity. RESULTS: Seventy-six patients were assessed. A P-PK-DDI with weak CYP3A4 or P-gp inhibition was found in 14 patients. In patients with P-PK-DDI or without, the dose was reduced during the first month in 57.1% and 17.7% (p = 0.003) and the drug withdrawn in 42.8% and 11.3% (p = 0.011), respectively. In multivariate analysis, a significant correlation was found between P-PK-DDI (CYP3A4 and P-gp inhibitors) and dose reduction, and between drug withdrawal and PK-DDI (CYP3A4 inhibitors). CONCLUSION: P-PK-DDI was correlated with dose reduction and drug withdrawal due to toxicity. The causality of this relationship warrants to be assessed; therefore, therapeutic drug monitoring is necessary in patients treated with TKI.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cytochrome P-450 CYP3A Inhibitors/toxicity , Pyrimidines/toxicity , Sulfonamides/toxicity , Sunitinib/toxicity , Aged , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , Indazoles , Male , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Retrospective Studies , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Sunitinib/administration & dosage , Sunitinib/pharmacokinetics
9.
Eur J Pharmacol ; 864: 172709, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31586633

ABSTRACT

Sunitinib is an oral small molecular tyrosine kinase inhibitor that exhibits potent antiangiogenic and antitumor activity. Unfortunately, sunitinib kidney toxicity limits its clinical use. Renal injury caused by sunitinib treatment can not only lead to the failure of cancer treatment, but also jeopardizes the health and life of patients. Currently, there is no better intervention measure for renal injury caused by sunitinib therapy except reducing the dosage or stopping the medication. In this study, we learned from clinical case report that sunitinib can cause severe renal injury. Subsequently, we compiled the clinical trials data of sunitinib found that sunitinib can cause general renal damage. Based on this finding, we conducted a study on the mechanism of sunitinib-induced renal injury. The results showed that sunitinib can inhibit the survival of HK-2 cells (human tubule epithelial cells) in a dose- and time-dependent manner. The survival inhibition is mainly due to the activation apoptotic signaling pathway by sunitinib in HK-2 cells and induces apoptosis of HK-2 cells. Subsequently, we found that natural compound oxypeucedanin can significantly alleviate the apoptosis of HK-2 cells induced by sunitinib. Through clinical investigation and experimental study of sunitinib, we found that sunitinib can cause extensive renal damage by inducing apoptosis of renal tubular epithelial cells and natural compound oxypeucedanin is a potentially effective intervention for nephrotoxicity of sunitinib. Thus, our research will provide a theoretical basis for the future rational use of sunitinib and the search for appropriate interventions for sunitinib-induced kidney damage.


Subject(s)
Kidney/cytology , Sunitinib/toxicity , Apoptosis/drug effects , Cell Line , Clinical Trials as Topic , Furocoumarins/pharmacology , Humans , Kidney/drug effects , Male , Middle Aged
10.
Arch Toxicol ; 93(11): 3249-3260, 2019 11.
Article in English | MEDLINE | ID: mdl-31552474

ABSTRACT

Sunitinib malate is a multi-targeted tyrosine kinase inhibitor used extensively for treatment of human tumors. However, cardiovascular adverse effects of sunitinib limit its clinical use. It is pivotal to elucidate molecular targets that mediate sunitinib-induced cardiotoxicity. Sirtuin 3 (Sirt3) is an effective mitochondrial deacetylase that has been reported to regulate sensitivity of different types of cells to chemotherapies, but roles of Sirt3 in sunitinib-induced cardiotoxicity have not been investigated. In the present study, we established wild type, Sirt3-knockout, and Sirt3-overexpressing mouse models of sunitinib (40 mg kg-1 day-1 for 28 days)-induced cardiotoxicity and examined cardiovascular functions and pathological changes. We further cultured wild type, Sirt3-knockout, and Sirt3-overexpressing primary mouse cardiac pericytes and analyzed sunitinib (10 µMol for 48 h)-induced alterations in cellular viability, cell death processes, and molecular pathways. Our results show that sunitinib predominantly induced hypertension, left ventricular systolic dysfunction, and cardiac pericyte death accompanied with upregulation of Sirt3 in cardiac pericytes, and these cardiotoxicities were significantly attenuated in Sirt3-knockout mice, but aggravated in Sirt3-overexpressing mice. Mechanistically, sunitinib induced cardiac pericyte death through inhibition of GSTP1/JNK/autophagy pathway and Sirt3 interacted with and inhibited GSTP1, further inhibiting the pathway and aggravating sunitinib-induced pericyte death. Conclusively, we demonstrate that Sirt3 promotes sensitivity to sunitinib-induced cardiotoxicity via GSTP1/JNK/autophagy pathway. Our results suggest Sirt3 might be a potential target for developing cardioprotective therapies for sunitinib-receiving patients.


Subject(s)
Antineoplastic Agents/toxicity , Glutathione S-Transferase pi/metabolism , Hypertension/chemically induced , MAP Kinase Kinase 4/metabolism , Pericytes/drug effects , Sirtuin 3/genetics , Sunitinib/toxicity , Animals , Autophagy/drug effects , Blood Pressure/drug effects , Cardiotoxicity , Cell Survival/drug effects , Heart/drug effects , Hypertension/metabolism , Hypertension/pathology , Mice , Mice, Knockout , Pericytes/metabolism , Pericytes/pathology , Signal Transduction
11.
Pharm Biol ; 57(1): 625-631, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31545912

ABSTRACT

Context: Sunitinib (SU) is a multi-targeted tyrosine kinase inhibitor anticancer agent whose clinical use is often limited by cardiovascular complications. Trimetazidine (TMZ) is an anti-angina agent that has been demonstrated cardioprotective effects in numerous cardiovascular conditions, but its potential effects in SU-induced cardiotoxicity have not been investigated. Objective: This study investigates the effect of TMZ in sunitinib-induced cardiotoxicity in vivo and in vitro and molecular mechanisms. Materials and methods: Male 129S1/SvImJ mice were treated with vehicle, SU (40 mg/kg/d) or SU and TMZ (20 mg/kg/d) via oral gavage for 28 days, and cardiovascular functions and cardiac protein expressions were examined. H9c2 cardiomyocytes were treated with vehicle, SU (2-10 µM) or SU and TMZ (40-120 µM) for 48 h, and cell viability, apoptosis, autophagy, and protein expression was tested. Results: SU induces hypertension (systolic blood pressure [SBP] + 28.33 ± 5.00 mmHg) and left ventricular dysfunction (left ventricular ejection fraction [LVEF] - 11.16 ± 2.53%) in mice. In H9c2 cardiomyocytes, SU reduces cell viability (IC50 4.07 µM) and inhibits the AMPK/mTOR/autophagy pathway (p < 0.05). TMZ co-administration with SU reverses SU-induced cardiotoxicity in mice (SBP - 23.75 ± 4.69 mmHg, LVEF + 10.95 ± 3.317%), alleviates cell viability loss in H9c2 cardiomyocytes (p < 0.01) and activates the AMPK/mTOR/autophagy pathway in vivo (p < 0.001) and in vitro (p < 0.05). Discussion and conclusions: Our results suggest TMZ as a potential cardioprotective approach for cardiovascular complications during SU regimen, and potentially for cardiotoxicity of other anticancer chemotherapies associated with cardiomyocyte autophagic pathways.


Subject(s)
Hypertension/drug therapy , Sunitinib/toxicity , Trimetazidine/pharmacology , AMP-Activated Protein Kinase Kinases , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cardiotoxicity , Cell Survival/drug effects , Cells, Cultured , Hypertension/chemically induced , Male , Mice , Microtubule-Associated Proteins , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Rats , Signal Transduction/drug effects , Sunitinib/pharmacology , TOR Serine-Threonine Kinases/metabolism
12.
Toxicology ; 426: 152281, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31445075

ABSTRACT

Sunitinib is cardiotoxic, but the mechanisms are not entirely clear. We aimed to enlarge our knowledge about the role of mitochondria in cardiac toxicity of sunitinib in vitro and in vivo. For this reason, we studied the toxicity of sunitinib on cardiac H9c2 cells exposed for 24 h, permeabilized rat cardiac fibers exposed for 15 min and in mice treated orally with sunitinib for 2 weeks (7.5 mg/kg/day). In H9c2 cells exposed for 24 h, sunitinib was more cytotoxic under galactose (favoring mitochondrial metabolism) compared to glucose conditions (favoring glycolysis). Sunitinib dissipated the mitochondrial membrane potential starting at 10 µM under glucose and at 5 µM under galactose conditions. Sunitinib reduced activities of mitochondrial enzyme complexes of the electron transport chain (ETC), increased mitochondrial ROS accumulation and decreased the cellular GSH pool. Electron microscopy revealed swollen mitochondria with loss of cristae. Accordingly, sunitinib caused caspase 3 activation and DNA fragmentation in H9c2 cells. Co-exposure with mito-TEMPO (mitochondrial-specific ROS scavenger) for 24 h prevented ATP and GSH depletion, as well as the increases in H2O2 and caspase 3/7 activity observed with sunitinib. In mice, treatment with sunitinib for two weeks increased plasma concentrations of troponin I and creatine kinase MB, indicating cardiomyocyte damage. The activity of enzyme complexes of the ETCwas decreased, mitochondrial ROS were increased and cleavage of caspase 3 was increased, suggesting cardiomyocyte apoptosis. In conclusion, mitochondrial damage with ROS accumulation appears to be an important mechanism of cardiotoxicity associated with sunitinib, eventually leading to apoptotic cell death.


Subject(s)
Antineoplastic Agents/toxicity , Heart Diseases/chemically induced , Mitochondria, Heart/drug effects , Oxidative Stress/drug effects , Sunitinib/toxicity , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Citrate (si)-Synthase/metabolism , DNA Fragmentation/drug effects , Electron Transport/drug effects , Enzyme Activation/drug effects , Heart Diseases/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria, Heart/enzymology
13.
Arch Toxicol ; 93(6): 1697-1712, 2019 06.
Article in English | MEDLINE | ID: mdl-31025080

ABSTRACT

Sunitinib (SNT) is a multi-targeted receptor tyrosine kinase inhibitor that has been approved by the FDA for cancer therapy. However, its cardiotoxicity has limited the clinical applicability with no effective therapeutic approach available. As a broadband kinase inhibitor, the function of several kinases that are essential to cardiac function might also be affected by SNT, such as calmodulin-dependent protein kinase (CaMKII), cyclic-AMP-dependent protein kinases (PKA), AMP-activated protein kinase (AMPK), and phosphoinositide 3 kinase (PI3K). In this study, we investigated whether SNT-induced cardiotoxicity could be prevented by blocking SNT-induced alteration in the corresponding signaling pathways. In human induced pluripotent stem cell-derived cardiomyocytes, SNT (0.5-20 µmol/L) inhibited contractility of cardiomyocytes in a concentration-dependent manner, and the inhibitory effect was prevented either by PIP3 (1 µmol/L) application or PI3K overexpression. On the contrary, the CaMKII inhibitor KN-93 (50 nmol/L), PKA inhibitor H89 (1 µmol/L), and AMPK activators metformin (2 mmol/L) and 5-aminoimidazole-4-carboxamide 1-b-D-ribofuranoside (2 mmol/L) presented negligible effects. Oral SNT administration (40 mg/kg/day) in mice progressively decreased the PI3K activity and cardiac function in 2 weeks with a significant decrease in the expression and activity of Cav1.2 and SERCA. Cardiac-specific PI3K overexpression through adeno-associated virus 9-mediated gene delivery in mice prevented SNT-induced reduction in cardiac function, calcium transient, calcium current, and Cav1.2 expression. In summary, our data indicate that increased PI3K activity is protective against SNT-induced calcium mishandling and contractile dysfunction. Cardiac-specific PI3K activation could be an effective therapeutic approach to treat SNT cardiotoxicity in patients with cancer.


Subject(s)
Antineoplastic Agents/toxicity , Heart Diseases/chemically induced , Heart Diseases/genetics , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sunitinib/toxicity , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Gene Transfer Techniques , Genetic Therapy , Heart Diseases/prevention & control , Humans , Induced Pluripotent Stem Cells , Myocytes, Cardiac/drug effects
14.
Toxicol In Vitro ; 58: 178-186, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30910525

ABSTRACT

The anti-cancer receptor tyrosine kinase inhibitors include known cardiotoxins: a component of this toxicity may be mediated by effects on cardiac fibroblasts (CFs). We hypothesised that imatinib mesylate (imatinib) and sunitinib malate (sunitinib) cause significant dysfunction in adult CFs. Following in vitro treatments with imatinib or sunitinib, adult rat CF viability was assessed by fluorescein diacetate assay, proliferation measured by bromodeoxyuridine nuclear incorporation and changes to the expression of CF secretome components determined by real time quantitative RT-PCR. Imatinib and sunitinib significantly reduced cell viability over 48 h, with EC50 values of 11.0 µM (imatinib) and 4.5 µM (sunitinib) respectively. Imatinib reduced CF proliferation from 35.5 ±â€¯3.2% in control to 23.0 ±â€¯5.5% (3 µM; p < 0.001) and to 9.4 ±â€¯2.5% (10 µM; p < 0.001), whereas sunitinib reduced proliferation to 22.9 ±â€¯3.1% (1 µM; p < 0.001) and to 15 ±â€¯1.0% (3 µM; p < 0.001). Further, 10 µM imatinib increased mRNA expression of TGFB1 7-fold, (p < 0.01), IL6 6-fold (p < 0.01), and IL1B 7-fold (p < 0.05) and reduced PDGFD 15-fold (p < 0.01); whereas sunitinib specifically reduced IL1B mRNA expression 17-fold (p < 0.01). Overall, these findings show tyrosine kinase inhibitors cause significant dysfunction in CFs. These data point to an important role for the PDGF pathway in governing CF functions, including survival and proliferation.


Subject(s)
Fibroblasts/drug effects , Imatinib Mesylate/toxicity , Protein Kinase Inhibitors/toxicity , Sunitinib/toxicity , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/genetics , Fibroblasts/metabolism , Male , Myocardium/cytology , Rats , Rats, Wistar
15.
Cardiovasc Toxicol ; 19(4): 344-356, 2019 08.
Article in English | MEDLINE | ID: mdl-30644033

ABSTRACT

This study has been initiated to investigate whether sunitinib (SUN) alters the expression of key genes engaged in mitochondrial transport and oxidation of long chain fatty acids (LCFA), and if so, whether these alterations should be viewed as a mechanism of SUN-induced cardiotoxicity, and to explore the molecular mechanisms whereby carnitine supplementation could attenuate SUN-induced cardiotoxicity. Adult male Wister albino rats were assigned to one of the four treatment groups: Rats in group 1 received no treatment but free access to tap water for 28 days. Rats in group 2 received L-carnitine (200 mg/kg/day) in drinking water for 28 days. Rats in group 3 received SUN (25 mg/kg/day) in drinking water for 28 days. Rats in group 4 received the same doses of L-carnitine and SUN in drinking water for 28 days. Treatment with SUN significantly increased heart weight, cardiac index, and cardiotoxicity enzymatic indices, as well as severe histopathological changes. Moreover, SUN significantly decreased level of adenosine monophosphate-activated protein kinase (AMPKα2), total carnitine, adenosine triphosphate (ATP) and carnitine palmitoyltransferase I (CPT I) expression and significantly increased acetyl-CoA carboxylase-2 (ACC2) expression and malonyl-CoA level in cardiac tissues. Interestingly, carnitine supplementation resulted in a complete reversal of all the biochemical, gene expression and histopathological changes-induced by SUN to the control values. In conclusion, data from this study suggest that SUN inhibits AMPK downstream signaling with the consequent inhibition of mitochondrial transport of LCFA and energy production in cardiac tissues. Carnitine supplementation attenuates SUN-induced cardiotoxicity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/toxicity , Carnitine/pharmacology , Dietary Supplements , Energy Metabolism/drug effects , Heart Diseases/prevention & control , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/toxicity , Sunitinib/toxicity , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Animals , Cardiotoxicity , Carnitine O-Palmitoyltransferase/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Male , Malonyl Coenzyme A/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Rats, Wistar , Signal Transduction
16.
Am J Physiol Heart Circ Physiol ; 316(3): H446-H458, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30499710

ABSTRACT

Although anticancer systemic therapy agents clearly lead to improved survival in patients with cancer, these can come at the cost of serious complications including cardiotoxicity. Two types of targeted systemic therapies currently in use for colorectal cancer (CRC) and renal cell cancer (RCC), respectively, include the vascular endothelial growth factor inhibitor bevacizumab (BVZ) and the tyrosine kinase inhibitor sunitinib (SNT). Despite the beneficial effects of BVZ and SNT in improving clinical outcomes in the settings of CRC and RCC, there is an increased risk of cardiac dysfunction. The aim of the present study was to determine whether prophylactic administration of renin-angiotensin system (RAS) inhibitors would attenuate the cardiotoxic side effects of BVZ or SNT in a chronic in vivo murine model. A total of 194 wild-type C57Bl/6 male mice received: 1) 0.9% saline, 2) BVZ (10 mg·kg-1·wk-1), or 3) SNT (40 mg·kg-1·day-1) for 4 wk. Within each arm, mice received daily prophylactic treatment with hydralazine (0.05 mg/ml), aliskiren (50 mg/kg), perindopril (4 mg/kg), or valsartan (2 mg/kg). Although hydralazine effectively lowered blood pressure in BVZ- or SNT-treated mice, it did not prevent left ventricular systolic dysfunction. Prophylactic administration of aliskiren, perindopril, or valsartan prevented adverse cardiovascular remodeling in mice treated with either BVZ or SNT. The addition of RAS antagonists also downregulated expression of phosphorylated p38 and Bcl-2-like 19-kDa interacting protein 3 in SNT-treated mice. In our chronic in vivo murine model, RAS antagonists partially attenuated the development of BVZ- or SNT-mediated cardiac dysfunction. Future clinical studies are warranted to investigate the cardioprotective effects of prophylactic treatment with RAS inhibitors in the settings of CRC and RCC. NEW & NOTEWORTHY In the evolving field of cardio-oncology, bevacizumab and sunitinib improve clinical outcomes in the settings of metastatic colorectal cancer and renal cell cancer, respectively. These anticancer drugs, however, are associated with an increased risk of cardiotoxicity. The prophylactic administration of renin-angiotensin system antagonists is partially cardioprotective against bevacizumab- and sunitinib-mediated cardiac dysfunction.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antihypertensive Agents/therapeutic use , Antineoplastic Agents/toxicity , Renin-Angiotensin System , Ventricular Dysfunction/prevention & control , Amides/administration & dosage , Amides/therapeutic use , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Bevacizumab/toxicity , Cardiotoxicity , Fumarates/administration & dosage , Fumarates/therapeutic use , Hydralazine/administration & dosage , Hydralazine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Perindopril/administration & dosage , Perindopril/therapeutic use , Sunitinib/toxicity , Valsartan/administration & dosage , Valsartan/therapeutic use , Ventricular Dysfunction/drug therapy , Ventricular Dysfunction/etiology
17.
Toxicology ; 411: 49-59, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30393206

ABSTRACT

Anti-cancer drug Sunitinib is linked to adverse cardiovascular events, which have shown to involve mitogen activated kinase kinase 7 (MKK7) pathway. Sunitinib-induced cardiotoxicity in 3, 12 and 24 months old male Sprague-Dawley rats and MKK7 expression and activation was investigated using the Langendorff perfused heart model followed by Western blot analysis. Cardiac function and infarct size were measured during/after 125 min of Sunitinib treatment. Left ventricular cardiac samples were analysed by qRT-PCR for expression of MKK7 mRNA and cardiac injury associated microRNAs. Infarct size was increased in all Sunitinib treated age groups. Haemodynamic alterations were observed following Sunitinib administration. Left ventricular developed pressure (LVDP) was decreased in all age groups, while heart rate (HR) was decreased in 3 and 12 months groups. Sunitinib treatment decreased the expression of miR-27a in all age groups, while miR-133a and miR-133b levels were increased in 3 months and decreased in 24 months groups. MKK7 mRNA and p-MKK7 levels were decreased in the 3 months group after Sunitinib treatment. MKK7 mRNA level was increased in 24 months group and p-MKK7 levels were increased in 12 months group following Sunitinib treatment. This study highlights the importance and impact of ageing and anti-cancer therapy-induced cardiotoxicity.


Subject(s)
Aging/physiology , Antineoplastic Agents/toxicity , Cardiotoxins/toxicity , Mitogen-Activated Protein Kinases/drug effects , Signal Transduction/drug effects , Sunitinib/toxicity , Animals , Heart Function Tests , Heart Rate/drug effects , Hemodynamics/drug effects , In Vitro Techniques , Male , MicroRNAs , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Ventricular Function, Left/drug effects
18.
J Am Heart Assoc ; 7(18): e009557, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30371202

ABSTRACT

Background The tyrosine kinase inhibitor sunitinib causes hypertension associated with reduced nitric oxide (NO) availability, elevated renal vascular resistance, and decreased fractional sodium excretion. We tested whether (1) nitrate supplementation mitigates sunitinib-induced hypertension and NO contributes less to renal vascular resistance as well as fractional sodium excretion regulation in sunitinib-treated rats than in controls; and (2) renal soluble guanylate cyclase (sGC) is downregulated and sGC activation lowers arterial pressure in rats with sunitinib-induced hypertension. Methods and Results Arterial pressure responses to nitrate supplementation and the effects of systemic and intrarenal NO synthase (NOS) inhibition on renal hemodynamics and fractional sodium excretion were assessed in sunitinib-treated rats and controls. Renal NOS and sGC mRNA as well as protein abundances were determined by quantitative polymerase chain reaction and Western blot. The effect of the sGC activator cinaciguat on arterial pressure was investigated in sunitinib-treated rats. Nitrate supplementation did not mitigate sunitinib-induced hypertension. Endothelium-dependent reductions in renal vascular resistance were similar in control and sunitinib-treated animals without and with systemic NOS inhibition. Selective intrarenal NOS inhibition lowered renal medullary blood flow in control but not in sunitinib-treated rats without significant effects on fractional sodium excretion. Renal cortical sGC mRNA and sGC α1-subunit protein abundance were less in sunitinib-treated rats than in controls, and cinaciguat effectively lowered arterial pressure by 15-20 mm Hg in sunitinib-treated rats. Conclusions Renal cortical sGC is downregulated in the presence of intact endothelium-dependent renal vascular resistance regulation in developing sunitinib-induced hypertension. This suggests that sGC downregulation occurs outside the renal vasculature, increases renal sodium retention, and contributes to nitrate resistance of sunitinib-induced hypertension.


Subject(s)
Blood Pressure/physiology , Down-Regulation , Guanylate Cyclase/metabolism , Hypertension/metabolism , Kidney/metabolism , Renal Circulation/physiology , Animals , Disease Models, Animal , Hypertension/chemically induced , Hypertension/physiopathology , Kidney/physiopathology , Male , Rats , Rats, Wistar , Sunitinib/toxicity , Vascular Resistance/physiology
19.
Biochem Pharmacol ; 155: 162-171, 2018 09.
Article in English | MEDLINE | ID: mdl-29983397

ABSTRACT

Tyrosine kinase inhibitors (TKIs) have advanced cancer treatment and prognosis but have also resulted in adverse effects such as fatigue, diarrhea, hypothyroidism, and other toxicities. We investigated TKI effects on skeletal muscle as a possible explanation of TKI induced fatigue. Changes in mitochondrial function due to inhibition of oxidative phosphorylation complexes, generation of superoxides, and inhibition of key transporters involved in uptake of glucose and/or nucleosides may result in alteration of energy metabolism and/or mitochondrial function. We investigated effects of imatinib, sorafenib and sunitinib on these processes in cultured C2C12 murine skeletal muscle cells. Imatinib, sorafenib and sunitinib were cytotoxic to C2C12 cells with IC50 values of 20, 8 and 8 µM, respectively. Imatinib stimulated glucose uptake and inhibited complex V activity by 35% at 50 µM. Sorafenib inhibited complex II/III and V with IC50 values of 32 and 28 µM, respectively. Sorafenib caused activation of caspase 3/7 and depolarization of mitochondrial membranes occurred very rapidly with complete loss at 5-10 µM. Sunitinib inhibited Complex I with an IC50 value of 38 µM and caused ATP depletion, caspase 3/7 activation, an increase in reactive oxygen species (ROS), and decreased nucleoside and glucose uptake. In conclusion, imatinib, sunitinib and sorafenib caused changes in mitochondrial complex activities, glucose and nucleoside uptake leading to decreased energy production and mitochondrial function in a skeletal muscle cell model, suggesting that these changes may play a role in fatigue, one of the most common adverse effects of TKIs.


Subject(s)
Energy Metabolism/drug effects , Imatinib Mesylate/toxicity , Muscle Fibers, Skeletal/drug effects , Protein Kinase Inhibitors/toxicity , Sorafenib/toxicity , Sunitinib/toxicity , Animals , Cell Line , Cells, Cultured , Cytotoxins/toxicity , Dose-Response Relationship, Drug , Energy Metabolism/physiology , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism
20.
Toxicology ; 409: 13-23, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30031043

ABSTRACT

Reports concerning hepatic mitochondrial toxicity of sunitinib are conflicting. We therefore decided to conduct a toxicological study in mice. After having determined the highest dose that did not affect nutrient ingestion and body weight, we treated mice orally with sunitinib (7.5 mg/kg/day) for 2 weeks. At the end of treatment, peak sunitinib plasma concentrations were comparable to those achieved in humans and liver concentrations were approximately 25-fold higher than in plasma. Sunitinib did not affect body weight, but increased plasma ALT activity 6-fold. The activity of enzyme complexes of the electron transport chain (ETC) was decreased numerically in freshly isolated and complex III activity significantly in previously frozen liver mitochondria. In previously frozen mitochondria, sunitinib decreased NADH oxidase activity concentration-dependently in both treatment groups. The hepatic mitochondrial reactive oxygen species (ROS) content and superoxide dismutase 2 expression were increased in sunitinib-treated mice. Protein and mRNA expression of several subunits of mitochondrial enzyme complexes were decreased in mitochondria from sunitinib-treated mice. Protein expression of PGC-1α, citrate synthase activity and mtDNA copy number were all decreased in livers of sunitinib-treated mice, indicating impaired mitochondrial proliferation. Caspase 3 activation and TUNEL-positive hepatocytes were increased in livers of sunitinib-treated mice, indicating hepatocyte apoptosis. In conclusion, sunitinib caused concentration-dependent toxicity in isolated mitochondria at concentrations reached in livers in vivo and inhibited hepatic mitochondrial proliferation. Daily mitochondrial insults and impaired mitochondrial proliferation most likely explain hepatocellular injury observed in mice treated with sunitinib.


Subject(s)
Antineoplastic Agents/toxicity , Hepatocytes/drug effects , Liver/drug effects , Mitochondria, Liver/drug effects , Protein Kinase Inhibitors/toxicity , Sunitinib/toxicity , Animals , Apoptosis/drug effects , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Hepatocytes/physiology , Liver/pathology , Liver/physiology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria, Liver/physiology , Necrosis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL