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










Base de dados
Intervalo de ano de publicação
1.
Clin Exp Pharmacol Physiol ; 49(1): 70-83, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34449914

RESUMO

Doxorubicin (DOX) is one of the most widely used chemotherapy agents; however, its nonselective effect causes cardiotoxicity. Curcumin (Cur), a well known dietary polyphenol, could exert a significant cardioprotective effect, but the biological application of this substance is limited by its chemical insolubility. To overcome this limitation, in this study, we synthesised gold nanoparticles based on Cur (Cur-AuNPs). Ultraviolet-visible (UV-Vis) absorbance spectroscopy and transmission electron microscopy (TEM) were performed for the characterisation of synthesised NPs, and Fourier transform infrared (FTIR) spectroscopy were applied to detect Cur on the surface of AuNPs. Its cytotoxicity effect on H9c2 cells was evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The biological efficacy of Cur-AuNPs was assessed after acute cardiotoxicity induction in BALB/c mice with DOX injection. The serum biomarkers, myocardial histological changes, and cardiomyocyte apoptosis were then measured. The results revealed that the heart protection by Cur-AuNPs is more effective than Cur alone. Heart protective effect of Cur-AuNPs was evident both in the short-term (24 hours) and long-term (14 days) study. The results of Cur-AuNPs400 after 24 hours of toxicity induction displayed the reduction of the cardiac injury serum biomarkers (LDH, CK-MB, cTnI, ADT, and ALT) and apoptotic proteins (Bax and Caspase-3), as well as increase of Bcl-2 anti-apoptotic proteins without any sign of interfibrillar haemorrhage and intercellular spaces in the heart tissue microscopic images. Our long-term study signifies that Cur-AuNPs400 in DOX-intoxicated mice could successfully inhibit body and heart weight loss in comparison to DOX group.


Assuntos
Apoptose/efeitos dos fármacos , Cardiotoxicidade/tratamento farmacológico , Cardiotoxinas/toxicidade , Curcumina/uso terapêutico , Doxorrubicina/toxicidade , Nanopartículas Metálicas , Animais , Cardiotoxicidade/etiologia , Cardiotoxinas/antagonistas & inibidores , Doxorrubicina/antagonistas & inibidores , Ouro , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Espectroscopia Fotoeletrônica , Espectroscopia de Infravermelho com Transformada de Fourier
2.
Pharmacol Res ; 169: 105642, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33933636

RESUMO

Anthracyclines including doxorubicin (DOX) are still the most widely used and efficacious antitumor drugs, although their cardiotoxicity is a significant cause of heart failure. Despite considerable efforts being made to minimize anthracycline-induced cardiac adverse effects, little progress has been achieved. In this study, we aimed to explore the role and underlying mechanism of SNX17 in DOX-induced cardiotoxicity. We found that SNX17 was downregulated in cardiomyocytes treated with DOX both in vitro and in vivo. DOX treatment combined with SNX17 interference worsened the damage to neonatal rat ventricular myocytes (NRVMs). Furthermore, the rats with SNX17 deficiency manifested increased susceptibility to DOX-induced cardiotoxicity (myocardial damage and fibrosis, impaired contractility and cardiac death). Mechanistic investigation revealed that SNX17 interacted with leiomodin-2 (LMOD2), a key regulator of the thin filament length in muscles, via its C-TERM domain and SNX17 deficiency exacerbated DOX-induced cardiac systolic dysfunction by promoting aberrant LMOD2 degradation through lysosomal pathway. In conclusion, these findings highlight that SNX17 plays a protective role in DOX-induced cardiotoxicity, which provides an attractive target for the prevention and treatment of anthracycline induced cardiotoxicity.


Assuntos
Cardiotoxinas/toxicidade , Doxorrubicina/toxicidade , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Nexinas de Classificação/metabolismo , Animais , Western Blotting , Cardiotoxinas/antagonistas & inibidores , Doxorrubicina/antagonistas & inibidores , Imunofluorescência , Células HEK293 , Humanos , Imunoprecipitação , Masculino , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Nexinas de Classificação/fisiologia
3.
Toxicology ; 441: 152508, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32525084

RESUMO

Doxorubicin (DOX) is one of the most effective and irreplaceable chemotherapeutic agents but its clinical use is limited due to its cardiotoxicity. Glycyrrhizin(GL) has been applied to liver disorders for long. However, little is known that if GL could be meaningful in attenuating cardiotoxicity. The aim of this study is to investigate the cardioprotective effects of GL in DOX-induced cardiotoxicity (DIC) and the underlying mechanism. Here, H9c2 cardiomyoblasts, Neonatal rat cardiomyocytes (NRCMs), and Rats were introduced as test models. A single dose of 20 mg/kg DOX (i.p.) was applied to induce acute cardiotoxicity in vivo, as reflected by growth inhibition, increased levels of AST and CK-MB, and reduction of SOD activity, while GL (25 or 50 mg/kg/d, 14 d, i.p.) could counteract these effects. Moreover, pre-incubation with GL (0.8 mM for 12 h) in H9c2 cells protected against DOX-induced cytotoxicity, oxidative stress and depolarization of mitochondrial membrane potential (MMP). Besides, Western blot analysis showed that DOX upregulated the expression of LC3 II and p62 whereas GL reversed that both in vitro and in vivo and improved the obstructed autophagy flux in DOX-treated H9c2 cells with an autophagy inhibitor Bafilomycin A1 (Baf A1, 50 nM, 2 h). It has been previously documented that High-mobility group box 1 (HMGB1) was involved in DIC. In our work, knockdown of HMGB1 significantly increased cell viability and LC3 II level in H9c2, suggesting HMGB1 was crucial in DOX-induced autophagy-triggering cell death. Intriguingly, GL is a direct inhibitor of HMGB1. We found that GL downregulated Akt/mTOR autophagy signaling pathway in DOX-treated H9c2 cells. More importantly, in non-silencing H9c2 cells (transfected with negative control siRNA) cells, the expression of phospho-Akt, phospho-mTOR, p62, and LC3 II was significantly decreased with GL pretreament compared to DOX alone. However, in H9c2/HMGB1-(transfected with HMGB1 siRNA) cells exposed to DOX, the expression of p-Akt, p-mTOR, p62, LC3 II had no statistical difference with or without GL, revealing that HMGB1 mediated the cardioprotective action of GL in DIC. Taken together, our findings demonstrate that improved autophagy flux via HMGB1-dependent Akt/mTOR signaling pathway might contribute to attenuate DIC and go a novel insight into the underlying mechanisms of GL's cardioprotective action. GL could be a potential candidate for the prevention of DIC.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Autofagia/efeitos dos fármacos , Cardiotoxinas/toxicidade , Doxorrubicina/toxicidade , Ácido Glicirrízico/farmacologia , Proteína HMGB1/metabolismo , Coração/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Western Blotting , Cardiotoxinas/antagonistas & inibidores , Linhagem Celular , Doxorrubicina/antagonistas & inibidores , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
4.
Clin Toxicol (Phila) ; 56(6): 389-396, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29069937

RESUMO

CONTEXT: High-dose insulin euglycaemia (HIE) is recommended in the management of toxin-induced cardiac toxicity, with increasing insulin doses now being used. We aimed to investigate the safety of HIE in toxin-induced cardiac toxicity. METHODS: This was a retrospective review of cases from two clinical toxicology units. Demographics, toxin(s) ingested, clinical effects, investigations (serum glucose, electrolytes), treatments (insulin, glucose, electrolyte replacement), length of stay (LOS) and outcomes were extracted from the patients' medical records. Associations between insulin and glucose/electrolyte homeostasis were explored by comparing insulin administration and glucose or electrolyte concentrations and replacement. RESULTS: There were 22 patients (12 females), median age 57 years (15-88 years) treated with HIE. There were 12 beta-blocker, six calcium channel blocker and three combined beta-blocker and calcium channel blocker ingestions. A total of 19 patients had a systolic blood pressure <80mmHg and 18 patients required inotropes in addition to HIE. There were three deaths. Despite glucose and electrolyte replacement, 16 patients (73%) developed hypoglycaemia (Reference range [RR] < 3.5 mmol/L or <63 mg/dl). In 7 patients, hypoglycaemia was mild (2.5-3.4 mmol/L or 45-62 mg/dl) and in nine was severe (<2.5 mmol/L or <45 mg/dl). There were no neurological effects from hypoglycaemia. A total of 18 patients (82%) developed hypokalaemia (<3.5 mEq/L). In 16 patients, this was mild (2.5-3.4 mEq/L). There were no cardiac arrhythmias associated with this hypokalaemia. There was no apparent association between insulin dosing and severity of hypoglycaemia or hypokalaemia, or in glucose or potassium replacement. Median insulin loading dose was 80U (range 50-125 U) and the median maximum insulin infusion rate was 150 U/h (range 38-1500 U/h). Median glucose infusions rates were 37.5g/h (range 4-75g/h). There was no apparent association between insulin and glucose administration. Glucose was administered for a median of 18h after ceasing insulin. The duration of glucose administration after ceasing insulin increased with the rate and total insulin administered during HIE. DISCUSSION: Despite the benefits of HIE in toxin-induced cardiac toxicity, it caused significant disruption to glucose and electrolyte homeostasis, although there were no apparent complications from this. There was no association by comparing the amount of insulin administered on adverse effects or glucose administered, suggesting higher doses of insulin are associated with no more adverse effects.


Assuntos
Cardiotoxicidade/tratamento farmacológico , Cardiotoxinas/intoxicação , Insulina/uso terapêutico , Adolescente , Antagonistas Adrenérgicos beta/intoxicação , Adulto , Idoso , Idoso de 80 Anos ou mais , Cardiotoxicidade/etiologia , Cardiotoxinas/antagonistas & inibidores , Feminino , Humanos , Insulina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Verapamil/intoxicação , Adulto Jovem
5.
Mar Drugs ; 12(5): 2922-36, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24828295

RESUMO

Echinochrome A (Ech A) is a naphthoquinoid pigment from sea urchins that possesses antioxidant, antimicrobial, anti-inflammatory and chelating abilities. Although Ech A is the active substance in the ophthalmic and cardiac drug Histochrome®, its underlying cardioprotective mechanisms are not well understood. In this study, we investigated the protective role of Ech A against toxic agents that induce death of rat cardiac myoblast H9c2 cells and isolated rat cardiomyocytes. We found that the cardiotoxic agents tert-Butyl hydroperoxide (tBHP, organic reactive oxygen species (ROS) inducer), sodium nitroprusside (SNP; anti-hypertension drug), and doxorubicin (anti-cancer drug) caused mitochondrial dysfunction such as increased ROS level and decreased mitochondrial membrane potential. Co-treatment with Ech A, however, prevented this decrease in membrane potential and increase in ROS level. Co-treatment of Ech A also reduced the effects of these cardiotoxic agents on mitochondrial oxidative phosphorylation and adenosine triphosphate level. These findings indicate the therapeutic potential of Ech A for reducing cardiotoxic agent-induced damage.


Assuntos
Cardiotônicos/farmacologia , Cardiotoxinas/antagonistas & inibidores , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Naftoquinonas/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Cardiotoxinas/toxicidade , Morte Celular/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Ouriços-do-Mar
6.
Toxicol Appl Pharmacol ; 276(1): 55-62, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24480152

RESUMO

Doxorubicin (Dox) is an indispensable chemotherapeutic agent for the treatment of various forms of neoplasia such as lung, breast, ovarian, and bladder cancers. Cardiotoxicity is a major concern for patients receiving Dox therapy. Previous work from our laboratory indicated that glucocorticoids (GCs) alleviate Dox-induced apoptosis in cardiomyocytes. Here we have found glucocorticoid-induced leucine zipper (GILZ) to be a mediator of GC-induced cytoprotection. GILZ was found to be induced in cardiomyocytes by GC treatment. Knocking down of GILZ using siRNA resulted in cancelation of GC-induced cytoprotection against apoptosis by Dox treatment. Overexpressing GILZ by transfection was able to protect cells from apoptosis induced by Dox as measured by caspase activation, Annexin V binding and morphologic changes. Western blot analyses indicate that GILZ overexpression prevented cytochrome c release from mitochondria and cleavage of caspase-3. When bcl-2 family proteins were examined, we found that GILZ overexpression causes induction of the pro-survival protein Bcl-xL. Since siRNA against Bcl-xL reverses GC induced cytoprotection, Bcl-xL induction represents an important event in GILZ-induced cytoprotection. Our data suggest that GILZ functions as a cytoprotective gene in cardiomyocytes.


Assuntos
Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Cardiotoxinas/antagonistas & inibidores , Doxorrubicina/antagonistas & inibidores , Glucocorticoides/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Antibióticos Antineoplásicos/efeitos adversos , Cardiotônicos/antagonistas & inibidores , Cardiotoxinas/efeitos adversos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Corticosterona/antagonistas & inibidores , Corticosterona/farmacologia , Doxorrubicina/efeitos adversos , Glucocorticoides/antagonistas & inibidores , Camundongos , Miócitos Cardíacos/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/agonistas , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Regulação para Cima/efeitos dos fármacos , Proteína bcl-X/agonistas , Proteína bcl-X/antagonistas & inibidores , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
7.
Clin Pharmacol Ther ; 95(1): 45-52, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24091715

RESUMO

Anthracyclines are powerful chemotherapy agents that are still widely used today. However, their clinical use is limited by the development of dose-dependent cardiotoxicity. Recently, we showed that topoisomerase 2ß (Top2ß) is required for anthracycline to induce DNA double-strand breaks and changes in the transcriptome, leading to mitochondrial dysfunction and generation of reactive oxygen species. Furthermore, deleting Top2ß from cardiomyocytes prevented the development of anthracycline-induced cardiotoxicity in mice. On the basis of this molecular insight, new strategies should be developed to prevent anthracycline-induced cardiotoxicity. First, Top2α-specific anthracyclines should be tested to determine whether they will spare the heart. Second, Top2ß should be studied as a potential biomarker to predict risk of developing cardiotoxicity before anthracycline treatment. Third, inhibiting and deleting Top2ß in the heart should also be tested as primary prevention strategies. We propose that Top2ß is a promising molecular target that can be used to design interventions to prevent anthracycline-induced cardiotoxicity.


Assuntos
Antraciclinas/toxicidade , Cardiotoxinas/toxicidade , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Marcação de Genes/métodos , Prevenção Primária/métodos , Animais , Antraciclinas/antagonistas & inibidores , Cardiotoxinas/antagonistas & inibidores , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/genética , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia
8.
Circulation ; 128(2): 152-61, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23757312

RESUMO

BACKGROUND: Doxorubicin (DOXO) is an effective anthracycline chemotherapeutic, but its use is limited by cumulative dose-dependent cardiotoxicity. Neuregulin-1ß is an ErbB receptor family ligand that is effective against DOXO-induced cardiomyopathy in experimental models but is also proneoplastic. We previously showed that an engineered bivalent neuregulin-1ß (NN) has reduced proneoplastic potential in comparison with the epidermal growth factor-like domain of neuregulin-1ß (NRG), an effect mediated by receptor biasing toward ErbB3 homotypic interactions uncommonly formed by native neuregulin-1ß. Here, we hypothesized that a newly formulated, covalent NN would be cardioprotective with reduced proneoplastic effects in comparison with NRG. METHODS AND RESULTS: NN was expressed as a maltose-binding protein fusion in Escherichia coli. As established previously, NN stimulated antineoplastic or cytostatic signaling and phenotype in cancer cells, whereas NRG stimulated proneoplastic signaling and phenotype. In neonatal rat cardiomyocytes, NN and NRG induced similar downstream signaling. NN, like NRG, attenuated the double-stranded DNA breaks associated with DOXO exposure in neonatal rat cardiomyocytes and human cardiomyocytes derived from induced pluripotent stem cells. NN treatment significantly attenuated DOXO-induced decrease in fractional shortening as measured by blinded echocardiography in mice in a chronic cardiomyopathy model (57.7±0.6% versus 50.9±2.6%, P=0.004), whereas native NRG had no significant effect (49.4±3.7% versus 50.9±2.6%, P=0.813). CONCLUSIONS: NN is a cardioprotective agent that promotes cardiomyocyte survival and improves cardiac function in DOXO-induced cardiotoxicity. Given the reduced proneoplastic potential of NN versus NRG, NN has translational potential for cardioprotection in patients with cancer receiving anthracyclines.


Assuntos
Cardiotônicos/farmacologia , Engenharia Química/métodos , Doxorrubicina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Neuregulina-1/genética , Neuregulina-1/farmacologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Cardiotoxinas/antagonistas & inibidores , Cardiotoxinas/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Doxorrubicina/antagonistas & inibidores , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Distribuição Aleatória , Ratos , Método Simples-Cego
9.
Pharm Biol ; 51(3): 339-44, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23134235

RESUMO

CONTEXT: Doxorubicin (Dox) is an anthracycline antibiotic used as anticancer agent. However, its use is limited due to its cardiotoxicity which is mainly attributed to accumulation of reactive oxygen species. OBJECTIVE: This study was conducted to assess whether the antioxidant, proanthocyanidins (Pro) can ameliorate Dox-induced cardiotoxicity in rats. MATERIALS AND METHODS: Male Sprague-Dawely rats were divided into four groups. Group I was control. Group II received Pro (70 mg/kg, orally) once daily for 10 days. Group III received doxorubicin 15 mg/kg i.p. as a single dose on the 7th day and Group IV animals were treated with Pro once daily for 10 days and Dox on the 7th day. The parameters of study were serum biomarkers, cardiac tissue antioxidant status, ECG, and effect on aconitine-induced cardiotoxicity. RESULTS: Cardiac toxicity of doxorubicin was manifested as a significant increase in heart rate, elevation of the ST segment, prolongation of the QT interval and an increase in T wave amplitude. In addition, Dox enhanced aconitine-induced cardiotoxicity by a significant decrease in the aconitine dose producing ventricular tachycardia (VT). Administration of Pro significantly suppressed Dox-induced ECG changes and normalized the aconitine dose producing VT. The toxicity of Dox was also confirmed biochemically by significant elevation of serum CK-MB and LDH activities as well as myocardial MDA and GSH contents and decrease in serum catalase and myocardial SOD activities. Administration of Pro significantly suppressed these biochemical changes. DISCUSSION AND CONCLUSION: These results suggest that proanthocyanidins might be a potential cardioprotective agent against Dox-induced cardiotoxicity due to its antioxidant properties.


Assuntos
Antioxidantes/uso terapêutico , Cardiomiopatia Dilatada/prevenção & controle , Cardiotônicos/uso terapêutico , Cardiotoxinas/antagonistas & inibidores , Doxorrubicina/antagonistas & inibidores , Extrato de Sementes de Uva/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Proantocianidinas/uso terapêutico , Aconitina/administração & dosagem , Aconitina/antagonistas & inibidores , Aconitina/toxicidade , Animais , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/antagonistas & inibidores , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/prevenção & controle , Biomarcadores/sangue , Cardiomiopatia Dilatada/induzido quimicamente , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , Cardiotoxinas/efeitos adversos , Doxorrubicina/efeitos adversos , Resistência a Medicamentos/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/fisiopatologia , Masculino , Miocárdio/metabolismo , Fitoterapia , Ratos , Ratos Sprague-Dawley , Taquicardia/induzido quimicamente , Taquicardia/prevenção & controle , Agonistas do Canal de Sódio Disparado por Voltagem/administração & dosagem , Agonistas do Canal de Sódio Disparado por Voltagem/toxicidade
10.
Can J Physiol Pharmacol ; 90(11): 1527-34, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23181280

RESUMO

Childhood cancer survivors can develop significant cardiac dysfunction in adulthood as a consequence of their cancer treatment. Studies have linked heart failure during pregnancy to childhood doxorubicin (DOX) exposure. We hypothesized that DOX injection would reduce cardiac function peripartum and that DOX-treated dams would show greater cardiac remodeling postweaning. Weanling female Sprague-Dawley rats were injected with phospate-buffered saline, DOX (3 mg/kg), or DOX plus the cardioprotectant dexrazoxane (DEX; 60 mg/kg) and followed for 2 pregnancies. DOX and DOX:DEX dams were fertile, but had fewer pups and more pup losses. Echocardiography, 1-day postpartum after each pregnancy, revealed greater increases in cardiac mass and eccentric hypertrophy in DOX-treated dams and early dilation in DOX:DEX dams. The expression of calcium homeostasis proteins can change after DOX treatment and cardiac remodeling. SERCA2a expression did not change. Reductions in phospholamban and phospho-serine 16-specific phospholamban expression in DOX dams were not relieved by DEX coinjection. DOX binds and inactivates calsequestrin 2 expression so increased calsequestrin 2 expression in DOX:DEX-treated dams suggests some DEX compensation. The eccentric hypertrophy and dilation development, despite compensatory changes in proteins controlling calcium cycling, suggest DOX damage with repeat pregnancy that was not alleviated fully by DEX.


Assuntos
Antineoplásicos/antagonistas & inibidores , Cardiotônicos/uso terapêutico , Doxorrubicina/antagonistas & inibidores , Ventrículos do Coração/efeitos dos fármacos , Complicações Cardiovasculares na Gravidez/prevenção & controle , Razoxano/uso terapêutico , Disfunção Ventricular Esquerda/prevenção & controle , Animais , Antineoplásicos/efeitos adversos , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/metabolismo , Calsequestrina/antagonistas & inibidores , Calsequestrina/biossíntese , Calsequestrina/metabolismo , Cardiotoxinas/efeitos adversos , Cardiotoxinas/antagonistas & inibidores , Doxorrubicina/efeitos adversos , Feminino , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Infertilidade Feminina/induzido quimicamente , Infertilidade Feminina/prevenção & controle , Período Pós-Parto , Gravidez , Complicações Cardiovasculares na Gravidez/induzido quimicamente , Complicações Cardiovasculares na Gravidez/metabolismo , Complicações Cardiovasculares na Gravidez/fisiopatologia , Resultado da Gravidez , Substâncias Protetoras/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Remodelação Ventricular/efeitos dos fármacos , Desmame
11.
Toxicology ; 292(1): 53-62, 2012 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-22155320

RESUMO

The long-term clinical usefulness of doxorubicin (DOX), an anthracycline with potent antitumor activity, is limited by DOX-induced cardiotoxicity. Kaempferol, one of the most common dietary flavonoids, is known to have anti-apoptotic, anti-oxidative, and anti-inflammatory properties. The current study aimed to investigate the possible protective effect of kaempferol against DOX-induced cardiotoxicity and the underlying mechanisms. Rats were intraperitoneally (i.p.) treated with DOX (3 mg/kg) every other day for a cumulative dose of 9 mg/kg. After 28 days, DOX caused retarded body and heart growth, oxidative stress, apoptotic damage, mitochondrial dysfunction, and Bcl-2 expression disturbance. In contrast, kaempferol pretreatment (10 mg/kg i.p. before DOX administration) attenuated the DOX-induced apoptotic damage in heart tissues. In vitro studies also indicated that kaempferol may have used the mitochondrion-dependent pathway to counteract the DOX-induced cardiotoxicity. This counteraction was achieved by inhibiting p53 expression and its binding to the promoter region of the Bax proapoptotic gene, but not to the Bcl-2 antiapoptotic gene. Kaempferol also effectively suppressed DOX-induced extracellular signal-regulated kinase (ERK) 1/2 activation, but had no effect on p38 and JNK. Therefore, kaempferol protected against DOX-induced cardiotoxicity, at least, partially, by inhibiting the activation of p53-mediated, mitochondrion-dependent apoptotic signaling, and by being involved in an ERK-dependent mitogen-activated protein kinase pathway. These findings elucidated the potential of kaempferol as a promising reagent for treating DOX-induced cardiotoxicity, and may have implications in the long-term clinical usefulness of DOX.


Assuntos
Cardiotônicos/farmacologia , Cardiotoxinas/toxicidade , Doxorrubicina/toxicidade , Quempferóis/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Animais , Cardiotônicos/química , Cardiotoxinas/antagonistas & inibidores , Células Cultivadas , Relação Dose-Resposta a Droga , Doxorrubicina/antagonistas & inibidores , Quempferóis/química , Masculino , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley
12.
J Int Med Res ; 39(5): 1720-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22117972

RESUMO

This study investigated the effects of berberine, a natural alkaloid, on doxorubicin-induced cardiotoxicity in mice. Mice were injected intraperitoneally with saline 10 ml/kg (n = 10), doxorubicin 2.5 mg/kg (n = 10), 60 mg/kg berberine 1 h before doxorubicin 2.5 mg/kg (n = 10), or 60 mg/kg berberine alone (n = 10) every other day for 14 days. Body weight, general condition and mortality were recorded over the 14-day study period. Electro cardiography was performed before the start of treatment and after 14 days and plasma lactate dehydrogenase (LDH) activity was measured after 14 days. At the end of the study period the heart was excised and examined histologically. An increase in mortality, an initial decrease in body weight, increased LDH activity, prolongation of QRS duration and increased myocardial injury were seen in the doxorubicin-treated group compared with the saline control group. These changes were significantly attenuated by pretreatment with berberine. The study suggests that berberine may have a potential protective role against doxorubicin-induced cardiotoxicity in mice.


Assuntos
Berberina/farmacologia , Cardiotônicos/farmacologia , Cardiotoxinas/antagonistas & inibidores , Doxorrubicina/antagonistas & inibidores , Animais , Eletrocardiografia , Feminino , L-Lactato Desidrogenase/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/patologia , Distribuição Aleatória , Aumento de Peso/efeitos dos fármacos
13.
Mol Cancer Ther ; 10(12): 2320-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21926192

RESUMO

AG014699 was the first inhibitor of the DNA repair enzyme PARP-1 to enter clinical trial in cancer patients. In addition to enhancing the cytotoxic effect of DNA-damaging chemotherapies, we have previously shown that AG014699 is vasoactive, thereby having the potential to improve drug biodistribution. The effectiveness of the clinical agent doxorubicin is confounded both by poor tumor penetration and cardiotoxicity elicited via PARP hyperactivation. In this study, we analyzed the impact of AG014699 on doxorubicin tolerance and response in breast (MDA-MB-231) and colorectal (SW620, LoVo) tumor models in vitro and in vivo. As anticipated, AG014699 did not potentiate the response to doxorubicin in vitro. In vivo, AG014699 did not influence the pharmacokinetics of doxorubicin; however, it did ameliorate cardiotoxicity. Both toxicity and extent of amelioration were more pronounced in male than in female mice. AG014699 improved vessel perfusion in both MDA-MB-231 and SW620 tumors; however, this neither led to improved tumor-accumulation of doxorubicin nor enhanced therapeutic response. In contrast, when combined with radiotherapy, AG014699 significantly enhanced response both in vitro and in vivo. Real-time assessment of tumor vessel function and companion histologic studies indicate that doxorubicin causes a profound antivascular effect that counters the positive effect of AG014699 on perfusion. These data indicate that although AG014699 can enhance response to some chemotherapeutic drugs via improved delivery, this does not apply to doxorubicin. PARP inhibitors may still be of use to counter doxorubicin toxicity, and if the gender effect translates from rodents to humans, this would have greater effect in males.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Indóis/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cardiotoxinas/antagonistas & inibidores , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Doxorrubicina/antagonistas & inibidores , Sinergismo Farmacológico , Feminino , Coração/efeitos dos fármacos , Humanos , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Perfusão , Inibidores de Poli(ADP-Ribose) Polimerases , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Toxicology ; 289(2-3): 122-31, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21864640

RESUMO

Catecholamines are stress hormones and sympathetic neurotransmitters essential for control of cardiac function and metabolism. However, pathologically increased catecholamine levels may be cardiotoxic by mechanism that includes iron-catalyzed formation of reactive oxygen species. In this study, five iron chelators used in clinical practice were examined for their potential to protect cardiomyoblast-derived cell line H9c2 from the oxidative stress and toxicity induced by catecholamines epinephrine and isoprenaline and their oxidation products. Hydroxamate iron chelator desferrioxamine (DFO) significantly reduced oxidation of catecholamines to more toxic products and abolished redox activity of the catecholamine-iron complex at pH 7.4. However, due to its hydrophilicity and large molecule, DFO was able to protects cells only at very high and clinically unachievable concentrations. Two newer chelators, deferiprone (L1) and deferasirox (ICL670A), showed much better protective potential and were effective at one or two orders of magnitude lower concentrations as compared to DFO that were within their clinically relevant plasma levels. Ethylenediaminetetraacetic acid (EDTA), dexrazoxane (ICRF-187, clinically approved cardioprotective agent against anthracycline-induced cardiotoxicity) as well as selected beta adrenoreceptor antagonists and calcium channel blockers exerted no effect. Hence, results of the present study indicate that small, lipophilic and iron-specific chelators L1 and ICL670A can provide significant protection against the oxidative stress and cardiomyocyte damage exerted by catecholamines and/or their reactive oxidation intermediates. This potential new application of the clinically approved drugs L1 and ICL670A warrants further investigation, preferably using more complex in vivo animal models.


Assuntos
Cardiotônicos/farmacologia , Cardiotoxinas/toxicidade , Catecolaminas/toxicidade , Quelantes de Ferro/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Cardiotoxinas/antagonistas & inibidores , Catecolaminas/antagonistas & inibidores , Linhagem Celular , Miócitos Cardíacos/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos
15.
Can J Physiol Pharmacol ; 87(8): 633-40, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19767888

RESUMO

Doxorubicin (DXR) is a chemotherapeutic agent used effectively in the treatment of several childhood malignancies. During treatment, cardiotoxicity caused by cell damage due to the free oxygen radicals that are generated is a major limiting factor. This study was undertaken to determine whether DXR-induced cardiotoxicity could be prevented by natural foods with antioxidant properties such as aged garlic extract (AGEX), grape seed proanthocyanidin (PA), and hazelnut. Wistar albino male rats were assigned randomly to 9 groups each consisting of 15 rats. AGEX, PA, and hazelnut groups received these antioxidants in addition to their standard rat diet. They were also treated with cumulative intraperitoneal (i.p.) injections according to 2 different regimens: either a high-dose of 15 mg/kg DXR (3.75 mg/kg per week for 4 weeks) or a low-dose of 7.5 mg/kg DXR (1.875 mg/kg per week for 4 weeks). The control group received i.p. 0.9% saline. AGEX, PA, or hazelnut supplements were given orally to the groups for a 6-week period starting 1 week before the DXR treatment and ending 1 week after the treatment. One week after the last DXR injection, heart tissue samples were analyzed to determine malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), and xanthine oxidase (XO) levels, and serum samples were taken for creatine kinase (CK). There were no significant changes in MDA levels among the control, DXR-treated groups, or supplemented groups that received additional natural antioxidant foods. SOD enzyme levels were decreased in rats treated with DXR. PA prevented the decrease at low doses of DXR. DXR treatment decreased CAT enzyme levels, but additional PA and hazelnut consumption increased these levels at low cumulative doses. XO enzyme levels were decreased in AGEX and hazelnut groups, but PA prevented the decrease. CK levels were elevated after DXR administration, indicating myocardial injury, but PA significantly reversed this. Although there were no differences histopathologically between AGEX, PA, and hazelnut groups, the protective effects of AGEX and PA were evident in electron microscopy. In conclusion, the positive effects of natural antioxidant foods on the prevention of DXR-induced cardiac injury could not be clearly shown on the basis of antioxidant enzymes. However, the electron microscopic changes clearly demonstrated the protective effects of AGEX and PA. The supplementation of these antioxidant foods over longer periods may show more definitive results. Human studies with different doses are needed to evaluate the effects of these foods on the human heart.


Assuntos
Corylus , Doxorrubicina/toxicidade , Alho , Extrato de Sementes de Uva/administração & dosagem , Cardiopatias/prevenção & controle , Extratos Vegetais/administração & dosagem , Proantocianidinas/administração & dosagem , Animais , Antioxidantes/uso terapêutico , Cardiotoxinas/antagonistas & inibidores , Cardiotoxinas/toxicidade , Doxorrubicina/antagonistas & inibidores , Cardiopatias/induzido quimicamente , Cardiopatias/dietoterapia , Mediadores da Inflamação/administração & dosagem , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/toxicidade , Masculino , Ratos , Ratos Wistar , Vitis
16.
Br J Cancer ; 101(5): 792-802, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19623174

RESUMO

BACKGROUND: Dexrazoxane (DEX, ICRF-187) is the only clinically approved cardioprotectant against anthracycline cardiotoxicity. It has been traditionally postulated to undergo hydrolysis to iron-chelating agent ADR-925 and to prevent anthracycline-induced oxidative stress, progressive cardiomyocyte degeneration and subsequent non-programmed cell death. However, the additional capability of DEX to protect cardiomyocytes from apoptosis has remained unsubstantiated under clinically relevant in vivo conditions. METHODS: Chronic anthracycline cardiotoxicity was induced in rabbits by repeated daunorubicin (DAU) administrations (3 mg kg(-1) weekly for 10 weeks). Cardiomyocyte apoptosis was evaluated using TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling) assay and activities of caspases 3/7, 8, 9 and 12. Lipoperoxidation was assayed using HPLC determination of myocardial malondialdehyde and 4-hydroxynonenal immunodetection. RESULTS: Dexrazoxane (60 mg kg(-1)) co-treatment was capable of overcoming DAU-induced mortality, left ventricular dysfunction, profound structural damage of the myocardium and release of cardiac troponin T and I to circulation. Moreover, for the first time, it has been shown that DEX affords significant and nearly complete cardioprotection against anthracycline-induced apoptosis in vivo and effectively suppresses the complex apoptotic signalling triggered by DAU. In individual animals, the severity of apoptotic parameters significantly correlated with cardiac function. However, this effective cardioprotection occurred without a significant decrease in anthracycline-induced lipoperoxidation. CONCLUSION: This study identifies inhibition of apoptosis as an important target for effective cardioprotection against chronic anthracycline cardiotoxicity and suggests that lipoperoxidation-independent mechanisms are involved in the cardioprotective action of DEX.


Assuntos
Antraciclinas/toxicidade , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Cardiotoxinas/toxicidade , Cardiopatias/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Razoxano/farmacologia , Animais , Antraciclinas/antagonistas & inibidores , Cardiotoxinas/antagonistas & inibidores , Cardiopatias/induzido quimicamente , Cardiopatias/patologia , Masculino , Miócitos Cardíacos/citologia , Coelhos
17.
Trends Pharmacol Sci ; 30(8): 403-10, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19595465

RESUMO

By interfering with signal transduction events that control cell proliferation and fate, kinase inhibitors (KIs) hold promise as anticancer agents. Nevertheless, the functional role of a kinase depends on the cellular context and hence kinase inhibition in off-target cells could lead to side effects. For instance, this context dependence renders many KIs potentially cardiotoxic since inhibition of primary cancer targets such as ABL, RAF1 or AMPK recruits pro-apoptotic pathways in cardiomyocytes. Motivated by these observations, we propose a mode of 'therapeutic editing' where one drug (the editor) suppresses the side effect promoted by the primary drug as it impacts off-target cells. Editor and primary drug have overlapping therapeutic impact, and the editor suppresses the downstream propagation of toxicity-related signaling.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/antagonistas & inibidores , Cardiotoxinas/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Humanos , Modelos Biológicos , Estrutura Molecular , Especificidade de Órgãos , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
18.
J Lipid Res ; 49(10): 2101-12, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18515784

RESUMO

Ceramide is among a number of potential lipotoxic molecules that are thought to modulate cellular energy metabolism. The heart is one of the tissues thought to become dysfunctional due to excess lipid accumulation. Dilated lipotoxic cardiomyopathy, thought to be the result of diabetes and severe obesity, has been modeled in several genetically altered mice, including animals with cardiac-specific overexpression of glycosylphosphatidylinositol (GPI)-anchored human lipoprotein lipase (LpL(GPI)). To test whether excess ceramide was implicated in cardiac lipotoxicity, de novo ceramide biosynthesis was inhibited pharmacologically by myriocin and genetically by heterozygous deletion of LCB1, a subunit of serine palmitoyltransferase (SPT). Inhibition of SPT, a rate-limiting enzyme in ceramide biosynthesis, reduced fatty acid and increased glucose oxidation in isolated perfused LpL(GPI) hearts, improved systolic function, and prolonged survival rates. Our results suggest a critical role for ceramide accumulation in the pathogenesis of lipotoxic cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Cardiotoxinas/metabolismo , Ceramidas/metabolismo , Animais , Biomarcadores/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Cardiotoxinas/antagonistas & inibidores , Bovinos , Ceramidas/antagonistas & inibidores , Ácidos Graxos/metabolismo , Ácidos Graxos Monoinsaturados/farmacologia , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Glicosilfosfatidilinositóis/metabolismo , Coração/efeitos dos fármacos , Coração/fisiopatologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Lipase Lipoproteica/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oxirredução , Fosforilação/efeitos dos fármacos , Serina C-Palmitoiltransferase/antagonistas & inibidores , Serina C-Palmitoiltransferase/genética , Serina C-Palmitoiltransferase/metabolismo , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...