Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Pharmacol Physiol ; 49(1): 70-83, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34449914

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxinas/toxicidad , Curcumina/uso terapéutico , Doxorrubicina/toxicidad , Nanopartículas del Metal , Animales , Cardiotoxicidad/etiología , Cardiotoxinas/antagonistas & inhibidores , Doxorrubicina/antagonistas & inhibidores , Oro , Masculino , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Espectroscopía de Fotoelectrones , Espectroscopía Infrarroja por Transformada de Fourier
2.
Biol Pharm Bull ; 44(11): 1732-1737, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34719649

RESUMEN

Doxorubicin (DOX) is an effective anticancer anthracycline drug; however, the cardiotoxicity limits its application. The aim of the present study was to investigate the potential protective effect of taurine against DOX-induced chronic cardiotoxicity in mice. We found that exogenous supplementation of taurine can inhibit the weight loss of mice caused by DOX. The increased activity of myocardial enzymes creatine kinase (CK) and lactate dehydrogenase (LDH) in response to DOX treatment were significantly hampered. In addition, taurine supplementation alleviated the decrease in superoxide dismutase (SOD) activity, glutathione (GSH) content, glutathione peroxidase 4 (Gpx4) expression, and the increase in malondialdehyde (MDA) content caused by DOX. Besides, taurine alleviated myocardial myofibrillar disruption and mitochondrial edema. Furthermore, our results showed that taurine decreased the expressions of cleaved caspase-3 and Bax/Bcl2, thereby inhibiting apoptosis. These collective data demonstrated that exogenous taurine supplementation has a potentially protective effect against the myocardial damage caused by doxorubicin in mice by enhancing antioxidant capacity and reducing oxidative damage and apoptosis.


Asunto(s)
Cardiotónicos/farmacología , Cardiotoxicidad/prevención & control , Cardiotoxinas/toxicidad , Doxorrubicina/toxicidad , Taurina/farmacología , Animales , Western Blotting , Caspasa 3/metabolismo , Glutatión/metabolismo , Ratones , Ratones Endogámicos ICR , Miocardio/enzimología , Miocardio/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Superóxido Dismutasa/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 41(3): 1019-1031, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33472401

RESUMEN

Drug-induced cardiotoxicity is a significant clinical issue, with many drugs in the market being labeled with warnings on cardiovascular adverse effects. Treatments are often prematurely halted when cardiotoxicity is observed, which limits their therapeutic potential. Moreover, cardiotoxicity is a major reason for abandonment during drug development, reducing available treatment options for diseases and creating a significant financial burden and disincentive for drug developers. Thus, it is important to minimize the cardiotoxic effects of medications that are in use or in development. To this end, identifying patients at a higher risk of developing cardiovascular adverse effects for the drug of interest may be an effective strategy. The discovery of human induced pluripotent stem cells has enabled researchers to generate relevant cell types that retain a patient's own genome and examine patient-specific disease mechanisms, paving the way for precision medicine. Combined with the rapid development of pharmacogenomic analysis, the ability of induced pluripotent stem cell-derivatives to recapitulate patient-specific drug responses provides a powerful platform to identify subsets of patients who are particularly vulnerable to drug-induced cardiotoxicity. In this review, we will discuss the current use of patient-specific induced pluripotent stem cells in identifying populations who are at risk to drug-induced cardiotoxicity and their potential applications in future precision medicine practice. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Cardiotoxicidad/etiología , Cardiotoxinas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Arritmias Cardíacas/inducido químicamente , Evaluación Preclínica de Medicamentos/métodos , Marcadores Genéticos , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/fisiología , Contracción Miocárdica/efectos de los fármacos , Miocarditis/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Pruebas de Farmacogenómica/métodos , Polimorfismo de Nucleótido Simple , Medicina de Precisión/métodos , Factores de Riesgo
4.
Toxicol In Vitro ; 67: 104891, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32446838

RESUMEN

Cardiotoxicity is an important toxicological endpoint for chemical and drug safety assessment. The present study aims to evaluate two stemcell-based in vitro models for cardiotoxicity screening of chemicals. Eleven model compounds were used to evaluate responses of mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) using beating arrest as a readout and the analysis of electrophysiological parameters measured with a multi-electrode array (MEA) platform of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Results revealed that the hiPSC-CM MEA assay responded to all compounds. The mESC-CM beating arrest assay was not responsive to potassium channel blockers and showed a lower sensitivity to sodium channel blockers and Na+/K+ ATPase inhibitors compared to the hiPSC-CM MEA assay. Calcium channel blockers and a ß-adrenergic receptor agonist showed comparable potencies in both models. The in vitro response concentrations from hiPSC-CMs were highly concordant with human effective serum concentrations of potassium and sodium channel blockers. It is concluded that both in vitro models enable the cardiotoxicity screening with different applicability domains. The mESC-CM beating arrest assay may be used as a first step in a tiered approach while the hiPSC-CM MEA assay may be the best starting point for quantitative in vitro to in vivo extrapolations.


Asunto(s)
Cardiotoxicidad , Cardiotoxinas/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/citología , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/efectos de los fármacos , Animales , Células Cultivadas , Ratones , Modelos Biológicos , Miocitos Cardíacos/fisiología
5.
Circ Res ; 125(9): 855-867, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31600125

RESUMEN

Given that cardiovascular safety concerns remain the leading cause of drug attrition at the preclinical drug development stage, the National Center for Toxicological Research of the US Food and Drug Administration hosted a workshop to discuss current gaps and challenges in translating preclinical cardiovascular safety data to humans. This white paper summarizes the topics presented by speakers from academia, industry, and government intended to address the theme of improving cardiotoxicity assessment in drug development. The main conclusion is that to reduce cardiovascular safety liabilities of new therapeutic agents, there is an urgent need to integrate human-relevant platforms/approaches into drug development. Potential regulatory applications of human-derived cardiomyocytes and future directions in employing human-relevant platforms to fill the gaps and overcome barriers and challenges in preclinical cardiovascular safety assessment were discussed. This paper is intended to serve as an initial step in a public-private collaborative development program for human-relevant cardiotoxicity tools, particularly for cardiotoxicities characterized by contractile dysfunction or structural injury.


Asunto(s)
Cardiotoxicidad/epidemiología , Cardiotoxinas/toxicidad , Educación/normas , Informe de Investigación/normas , United States Food and Drug Administration/normas , Animales , Cardiotoxicidad/prevención & control , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Evaluación Preclínica de Medicamentos/tendencias , Educación/tendencias , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Informe de Investigación/tendencias , Estados Unidos/epidemiología , United States Food and Drug Administration/tendencias
6.
Toxicol Appl Pharmacol ; 383: 114785, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31629732

RESUMEN

Celastrol (CS), an active triterpene derived from traditional Chinese medicine Tripterygium wilfordii Hook. f, has been used to treat chronic inflammation, arthritis and other diseases. However, it has been reported that CS can trigger cardiotoxicity and the molecular mechanism of heart injury induced by CS is not clear. Considering the wide application of Tripterygium wilfordii Hook. f in clinics, it is necessary to develop an accurate and reliable method to assess the safety of CS, and to elucidate as much as possible the mechanism of cardiotoxicity induced by CS. In this study, Ultra-performance liquid chromatography coupled with quadrupole time of flight mass spectrometry (UPLC-Q-TOF/MS)-based metabolomics revealed clues to the mechanism of CS-induced heart injury. Palmitic acid significantly increased in plasma from CS-treated rats, and this increase resulted in oxidative stress response in vivo. Excessive ROS further activate TNF signaling pathway and caspase family, which were obtained from the KEGG enrichment analysis of network toxicology strategy. Protein expression level of caspase-3, caspase-8, bax were significantly increased by western blot. Q-PCR also showed the similar results as western blot. It means that apoptosis plays a key role in the process of celastrol induced cardiotoxicity. Blocking this signal axis may be a potential way to protect myocardial tissue.


Asunto(s)
Cardiotoxinas/toxicidad , Redes y Vías Metabólicas/efectos de los fármacos , Metabolómica/métodos , Tripterygium/toxicidad , Triterpenos/toxicidad , Animales , Cardiotoxicidad/metabolismo , Cardiotoxinas/metabolismo , Masculino , Redes y Vías Metabólicas/fisiología , Triterpenos Pentacíclicos , Ratas , Ratas Wistar , Tripterygium/metabolismo , Triterpenos/metabolismo
7.
Biotechnol Adv ; 36(8): 2232-2247, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30342084

RESUMEN

With unique characteristics such as high surface area, capacity of various functionalization, low weight, high conductivity, thermal and chemical stability, and free radical scavenging, carbon nanomaterials (CNMs) such as carbon nanotubes (CNTs), fullerene, graphene (oxide), carbon nanohorns (CNHs), and their derivatives have increasingly been utilized in nanomedicine and biomedicine. On the one hand, owing to ever-increasing applications of CNMs in technological and industrial fields as well as presence of combustion-derived CNMs in the ambient air, the skepticism has risen over the adverse effects of CNMs on human being. The influences of CNMs on cardiovascular system and cardiovascular diseases (CVDs) such as atherosclerosis, of which consequences are ischemic heart disease and ischemic stroke, as the main causes of death, is of paramount importance. In this regard, several studies have been devoted to specify the biomedical applications and cardiovascular toxicity of CNMs. Therefore, the aim of this review is to specify the roles and applications of various CNMs in atherosclerosis, and also identify the key role playing parameters in cardiovascular toxicity of CNMs so as to be a clue for prospective deployment of CNMs.


Asunto(s)
Aterosclerosis , Carbono/toxicidad , Cardiotoxinas/toxicidad , Nanoestructuras/toxicidad , Animales , Aterosclerosis/inducido químicamente , Aterosclerosis/inmunología , Aterosclerosis/fisiopatología , Fulerenos/toxicidad , Humanos , Ratones , Nanomedicina
8.
J Basic Clin Physiol Pharmacol ; 30(1): 73-79, 2018 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-30110251

RESUMEN

Background Because of the antioxidant effects of Zataria multiflora (ZM) and carvacrol (CAR) and also the role of oxidative stress in the induction of cardiotoxicity induced by Adriamycin (ADR), the aim of this study was to investigate the improvement effects of ZM extract and CAR on cardiotoxicity induced by ADR in rats. Methods Twenty-eight male rats were randomly assigned to four groups including (1) the control group; (2) the ADR group, which received ADR intravenously at the beginning of the study and the (3) ZM+ADR and (4) CAR+ADR groups, which received ZM and CAR by gavage for 28 consecutive days and ADR as single dose. Blood samples were collected on days 0 and 28 to determine serum glutamic oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT) and lactate dehydrogenase (LDH). Also, cardiac tissue was removed for redox marker evaluation. Results In the ADR group, malondialdehyde (MDA) significantly increased and superoxide dismutase (SOD) activity and total thiol contents significantly reduced, as compared with the control group, while CAR administration significantly improved this condition. Treatment with ZM significantly increased the SOD activity and total thiol content, as compared with the ADR group. The level of LDH significantly increased on day 28 in the ADR group compared to the control group, and administration of ZM and CAR significantly decreased it. The SGPT and SGOT levels in the ADR group significantly increased, and CAR administration significantly reduced them. Conclusion The results indicate that the administration of ZM hydroalcoholic extracts and its active ingredient, CAR, could reduce the oxidative stress damage through promotion of the cardiac and systemic antioxidant system. Also, CAR administration demonstrated better improvement in cardiotoxicity with ADR in rats.


Asunto(s)
Cardiotoxinas/toxicidad , Doxorrubicina/toxicidad , Lamiaceae , Monoterpenos/farmacología , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Antibióticos Antineoplásicos/toxicidad , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacología , Cimenos , Masculino , Estrés Oxidativo/fisiología , Extractos Vegetales/aislamiento & purificación , Distribución Aleatoria , Ratas , Ratas Wistar
9.
Toxicol Mech Methods ; 28(7): 488-498, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29564938

RESUMEN

Acrolein is a reactive electrophilic aldehyde known to cause mitochondrial dysfunction, oxidative stress, and dysregulation of signaling transduction in vitro. Most in vitro systems employ standard cell culture maintenance conditions of 95% air/5% CO2, translating to a culture oxygen tension of approximately 20%, far above most physiological tissues. The purpose of this investigation was to examine whether low-serum, retinoic acid differentiated H9c2 cells were less sensitive to acrolein insult when cultured under reduced oxygen tension. H9c2 cells were maintained separately in 20% and 5% oxygen, differentiated for 5 d, and then exposed to acrolein for 30 min in media containing varying concentrations of tricarboxylic acid and glycolytic substrates, followed by fresh medium replacement. Cells were then assessed for MTT reduction at 2 h and 24 h after acrolein insult. We showed that pyruvate supplementation in combination with lowered oxygen culturing significantly attenuated acrolein-induced viability loss at 24 h. Poly(ADP-ribose) polymerase inhibition and EGTA preferentially provided partial rescue to low oxygen cultures, but not for standard cultures. Collectively, these results offer evidence supporting altered toxicogenic response of H9c2 during physiologically relevant oxygen tension culturing.


Asunto(s)
Acroleína/toxicidad , Cardiotoxinas/toxicidad , Mioblastos Cardíacos/efectos de los fármacos , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Ácido Pirúvico/metabolismo , Animales , Antioxidantes/farmacología , Biomarcadores/metabolismo , Quelantes del Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Medicamentos , Glucólisis/efectos de los fármacos , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Ratas , Pruebas de Toxicidad Aguda , Ácidos Tricarboxílicos/metabolismo
10.
BMC Complement Altern Med ; 17(1): 554, 2017 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-29284479

RESUMEN

BACKGROUND: The use of doxorubicin (DOX) an anthracycline antineoplastic agent is withdrawn due to its cardio-toxic side effects. Oxidative stress has been recognized as the primary cause of DOX induced cardiotoxicity. We have investigated whether polyphenol rich ethyl acetate extract of Acacia hydaspica (AHE) can attenuate doxorubicin-induced cardiotoxicity via inhibition of oxidative stress. METHODS: AHE was administered orally to rats once daily for 6 weeks at doses of 200 and 400 mg/kg b.w. DOX (3 mg/kg b.w. i.p., single dose/week) was administered for 6 weeks (chronic model). The parameters studied to evaluate cardioprotective potential were the serum cardiac function biomarkers (CK, CKMB, AST and LDH), hematological parameters, cardiac tissue antioxidant enzymatic status and oxidative stress markers, and histopathological analysis to validate biochemical findings. RESULTS: Chronic 6 week treatment of DOX significantly deteriorated cardiac function biomarkers and decreased the activities of antioxidant enzymes, whereas significant increase in oxidative stress biomarkers was noticed in comparison to control group. AHE dose dependently protected DOX-induced leakage of cardiac enzymes in serum and ameliorated DOX-induced oxidative stress; as evidenced by decreasing lipid peroxidation, H2O2 and NO content with increase in phase I and phase II antioxidant enzymes. Doxorubicin treatment produced severe morphological lesions, leucopenia, decrease in red blood cell counts and hemoglobin concentrations. AHE co-treatment protected the heart and blood elements from the toxic effects of doxorubicin as indicated by the recovery of hematological parameters to normal values and prevention of myocardial injuries in a dose dependent way. The protective potency of AHE (400 mg/kg b.w) was equivalent to silymarin. CONCLUSION: Results revealed that AHE showed protective effects against DOX induce cardiotoxicity. The protective effect might attribute to its polyphenolic constituents and antioxidant properties. AHE might be helpful in combination therapies as safer and efficient.


Asunto(s)
Acacia/química , Antioxidantes/farmacología , Cardiotoxinas/toxicidad , Doxorrubicina/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Antioxidantes/química , Biomarcadores/sangre , Corazón/efectos de los fármacos , Masculino , Extractos Vegetales/química , Polifenoles/química , Polifenoles/farmacología , Ratas , Ratas Sprague-Dawley
11.
Biochem Biophys Res Commun ; 494(1-2): 138-143, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29042197

RESUMEN

Microelectrode array (MEA) based-drug screening with human induced pluripotent stem cell-derived cardiomyocytes (hiPSCM) is a potent pre-clinical assay for efficiently assessing proarrhythmic risks in new candidates. Furthermore, predicting sympathetic modulation of the proarrhythmic side-effects is an important issue. Although we have previously developed an MEA-based co-culture system of rat primary cardiomyocyte and sympathetic neurons (rSNs), it is unclear if this co-culture approach is applicable to develop and investigate sympathetic innervation of hiPSCMs. In this study, we developed a co-culture of rSNs and hiPSCMs on MEA substrate, and assessed functional connections. The inter-beat interval of hiPSCM was significantly shortened by stimulation in SNs depending on frequency and pulse number, indicating functional connections between rSNs and hiPSCM and the dependency of chronotropic effects on rSN activity pattern. These results suggest that our co-culture approach can evaluate sympathetic effects on hiPSCMs and would be a useful tool for assessing sympathetic modulated-cardiotoxicity in human cardiac tissue.


Asunto(s)
Técnicas de Cocultivo/instrumentación , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/fisiología , Neuronas/fisiología , Animales , Arritmias Cardíacas/inducido químicamente , Cardiotoxinas/toxicidad , Células Cultivadas , Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Estimulación Eléctrica , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Microelectrodos , Miocitos Cardíacos/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas
12.
Biomed Chromatogr ; 31(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28544073

RESUMEN

The acute cardiotoxicity induced by Veratrum nigrum (VN) is explored by analyzing heart tissue metabolic profiles in mouse models and applying reversed-phase liquid chromatography mass spectrometry and hydrophilic interaction liquid chromatography mass spectrometry that are based on ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry. An animal model of acute heart injury was established in mice via intra-gastric administration of VN. Then, electrocardiogram and echocardiograph monitoring of cardiac function and pathological examination were performed on mice in both the control and VN groups, and it was verified that acute heart injury was caused. Meanwhile, comparing the results of the control and VN groups, we detected 36 differential endogenous metabolites of heart tissue, including taurine, riboflavin, purine and lipids, which are related to many possible pathways such as purine metabolism, taurine and hypotaurine metabolism and energy metabolism. Our study provides a scientific approach for evaluating and revealing the mechanisms of VN-induced cardiotoxicity via the metabolomic strategy.


Asunto(s)
Cardiotoxinas/toxicidad , Cromatografía Líquida de Alta Presión/métodos , Metaboloma/efectos de los fármacos , Extractos Vegetales/toxicidad , Veratrum/química , Animales , Cardiotoxicidad/metabolismo , Cardiotoxinas/química , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Espectrometría de Masas/métodos , Redes y Vías Metabólicas , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Extractos Vegetales/química
13.
Int J Mol Sci ; 17(8)2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27490540

RESUMEN

Endothelin-1 (ET-1) autocrine and paracrine signaling modulate cell proliferation of tumor cells by activating its receptors, endothelin A receptor (ETAR) and endothelin B receptor (ETBR). Dysregulation of ETAR activation promotes tumor development and progression. The potential of ETAR antagonists and the dual-ETAR and ETBR antagonists as therapeutic approaches are under preclinical and clinical studies. Salvianolic acid A (Sal A) is a hydrophilic polyphenolic derivative isolated from Salvia miltiorrhiza Bunge (Danshen), which has been reported as an anti-cancer and cardio-protective herbal medicine. In this study, we demonstrate that Sal A inhibits ETAR activation induced by ET-1 in both recombinant and endogenous ETAR expression cell lines. The IC50 values were determined as 5.7 µM in the HEK293/ETAR cell line and 3.14 µM in HeLa cells, respectively. Furthermore, our results showed that Sal A suppressed cell proliferation and extended the doubling times of multiple cancer cells, including HeLa, DU145, H1975, and A549 cell lines. In addition, Sal A inhibited proliferation of DU145 cell lines stimulated by exogenous ET-1 treatment. Moreover, the cytotoxicity and cardio-toxicity of Sal A were assessed in human umbilical vein endothelial cells (HUVEC) and Human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs), which proved that Sal A demonstrates no cytotoxicity or cardiotoxicity. Collectively, our findings indicate that Sal A is a novel anti-cancer candidate through targeting ETAR.


Asunto(s)
Ácidos Cafeicos/farmacología , Antagonistas de los Receptores de la Endotelina A/farmacología , Lactatos/farmacología , Neoplasias/patología , Receptor de Endotelina A/metabolismo , Cardiotoxinas/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Reproducibilidad de los Resultados
14.
Nat Commun ; 7: 11542, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27161419

RESUMEN

The improvement of preclinical cardiotoxicity testing, discovery of new ion-channel-targeted drugs, and phenotyping and use of stem cell-derived cardiomyocytes and other biologics all necessitate high-throughput (HT), cellular-level electrophysiological interrogation tools. Optical techniques for actuation and sensing provide instant parallelism, enabling contactless dynamic HT testing of cells and small-tissue constructs, not affordable by other means. Here we show, computationally and experimentally, the limits of all-optical electrophysiology when applied to drug testing, then implement and validate OptoDyCE, a fully automated system for all-optical cardiac electrophysiology. We validate optical actuation by virally introducing optogenetic drivers in rat and human cardiomyocytes or through the modular use of dedicated light-sensitive somatic 'spark' cells. We show that this automated all-optical approach provides HT means of cellular interrogation, that is, allows for dynamic testing of >600 multicellular samples or compounds per hour, and yields high-content information about the action of a drug over time, space and doses.


Asunto(s)
Técnicas Electrofisiológicas Cardíacas/métodos , Optogenética/métodos , Animales , Automatización , Cardiotoxinas/toxicidad , Células Cultivadas , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Nifedipino/administración & dosificación , Nifedipino/toxicidad , Ratas
15.
Arch Toxicol ; 90(12): 3087-3098, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26842497

RESUMEN

An in depth investigation at the genomic level is needed to identify early human-relevant cardiotoxicity biomarkers that are induced by drugs and environmental toxicants. The main objective of this study was to investigate the role of microRNAs (miRNAs) as cardiotoxicity biomarkers using human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) that were exposed to doxorubicin (DOX) as a "gold standard" cardiotoxicant. hiPSC-CMs were exposed to 156 nM DOX for 2 days or for 6 days of repeated exposure, followed by drug washout and incubation in drug-free culture medium up to day 14 after the onset of exposure. The induced miRNAs were profiled using miRNA microarrays, and the analysis of the data was performed using the miRWalk 2.0 and DAVID bioinformatics tools. DOX induced early deregulation of 14 miRNAs (10 up-regulated and 4 down-regulated) and persistent up-regulation of 5 miRNAs during drug washout. Computational miRNA gene target predictions suggested that several DOX-responsive miRNAs might regulate the mRNA expression of genes involved in cardiac contractile function. The hiPSC-CMs exposed to DOX in a range from 39 to 156 nM did not show a significant release of the cytotoxicity marker lactate dehydrogenase (LDH) compared to controls. Quantitative real-time PCR analyses confirmed the early deregulation of miR-187-3p, miR-182-5p, miR-486-3p, miR-486-5p, miR-34a-3p, miR-4423-3p, miR-34c-3p, miR-34c-5p and miR-1303, and also the prolonged up-regulation of miR-182-5p, miR-4423-3p and miR-34c-5p. Thus, we identified and validated miRNAs showing differential DOX-responsive expression before the occurrence of cytotoxicity markers such as LDH, and these miRNAs also demonstrated the significant involvement in heart failure in patients and animal models. These results suggest that the DOX-induced deregulated miRNAs in human CMs may be used as early sensitive cardiotoxicity biomarkers for screening potential drugs and environmental cardiotoxicants with a similar mechanism of action.


Asunto(s)
Cardiotoxinas/toxicidad , Doxorrubicina/toxicidad , MicroARNs/metabolismo , Modelos Químicos , Miocitos Cardíacos/efectos de los fármacos , Biomarcadores/metabolismo , Biomarcadores Farmacológicos/metabolismo , Diferenciación Celular , Células Cultivadas , Biología Computacional , Evaluación Preclínica de Medicamentos , Drogas en Investigación/efectos adversos , Monitoreo del Ambiente , Contaminantes Ambientales/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Cinética , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Toxicol Appl Pharmacol ; 296: 42-53, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26821276

RESUMEN

The recent establishment of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), which express the major cardiac ion channels and recapitulate spontaneous mechanical and electrical activities, may provide a possible solution for the lack of in vitro human-based cardiotoxicity testing models. Cardiotoxicity induced by the antidepressant nefazodone was previously revealed to cause an acquired QT prolongation by hERG channel blockade. To elucidate the cellular mechanisms underlying the cardiotoxicity of nefazodone beyond hERG, its effects on cardiac action potentials (APs) and ion channels were investigated using hiPSC-CMs with whole-cell patch clamp techniques. In a proof of principle study, we examined the effects of cardioactive channel blockers on the electrophysiological profile of hiPSC-CMs in advance of the evaluation of nefazodone. Nefazodone dose-dependently prolonged the AP duration at 90% (APD90) and 50% (APD50) repolarization, reduced the maximum upstroke velocity (dV/dtmax) and induced early after depolarizations. Voltage-clamp studies of hiPSC-CMs revealed that nefazodone inhibited various voltage-gated ion channel currents including IKr, IKs, INa, and ICa. Among them, IKr and INa showed relatively higher sensitivity to nefazodone, consistent with the changes in the AP parameters. In summary, hiPSC-CMs enabled an integrated approach to evaluate the complex interactions of nefazodone with cardiac ion channels. These results suggest that hiPSC-CMs can be an effective model for detecting drug-induced arrhythmogenicity beyond the current standard assay of heterologously expressed hERG K(+) channels.


Asunto(s)
Antidepresivos de Segunda Generación/toxicidad , Cardiotoxinas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Triazoles/toxicidad , Animales , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , Evaluación Preclínica de Medicamentos/métodos , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/fisiología , Miocitos Cardíacos/fisiología , Piperazinas , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley
17.
PLoS One ; 10(3): e0119515, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25781638

RESUMEN

Venenum Bufonis, a well-known traditional Chinese medicine, has been widely used in Asia and has gained popularity in Western countries over the last decade. Venenum Bufonis has obvious side effects that have been observed in clinical settings, but few studies have reported on its cardiotoxicity. In this work, the cardiotoxicity of Venenum Bufonis was investigated using a 11H NMR-based metabolomics approach. The 1H NMR profiles of the serum, myocardial extracts and liver extracts of specific-pathogen-free rats showed that Venenum Bufonis produced significant metabolic perturbations dose-dependently with a distinct time effect, peaking at 2 hr after dosing and attenuating gradually. Clinical chemistry, electrocardiographic recordings, and histopathological evaluation provided additional evidence of Venenum Bufonis-induced cardiac damage that complemented and supported the metabolomics findings. The combined results demonstrated that oxidative stress, mitochondrial dysfunction, and energy metabolism perturbations were associated with the cardiac damage that results from Venenum Bufonis.


Asunto(s)
Bufanólidos/toxicidad , Cardiotoxinas/toxicidad , Metaboloma/efectos de los fármacos , Miocardio/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Espectroscopía de Resonancia Magnética , Masculino , Miocardio/patología , Ratas
19.
J Control Release ; 198: 62-70, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25483426

RESUMEN

Triglyceride micro-emulsions such as Intralipid® have been used to reverse cardiac toxicity induced by a number of drugs but reservations about their broad-spectrum applicability remain because of the poorly understood mechanism of action. Herein we report an integrated mechanism of reversal of bupivacaine toxicity that includes both transient drug scavenging and a cardiotonic effect that couple to accelerate movement of the toxin away from sites of toxicity. We thus propose a multi-modal therapeutic paradigm for colloidal bio-detoxification whereby a micro-emulsion both improves cardiac output and rapidly ferries the drug away from organs subject to toxicity. In vivo and in silico models of toxicity were combined to test the contribution of individual mechanisms and reveal the multi-modal role played by the cardiotonic and scavenging actions of the triglyceride suspension. These results suggest a method to predict which drug toxicities are most amenable to treatment and inform the design of next-generation therapeutics for drug overdose.


Asunto(s)
Bupivacaína/toxicidad , Cardiotónicos/uso terapéutico , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxinas/toxicidad , Fosfolípidos/uso terapéutico , Aceite de Soja/uso terapéutico , Triglicéridos/uso terapéutico , Animales , Bupivacaína/farmacocinética , Gasto Cardíaco/efectos de los fármacos , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacología , Cardiotoxicidad/metabolismo , Cardiotoxicidad/fisiopatología , Cardiotoxinas/farmacocinética , Emulsiones/administración & dosificación , Emulsiones/farmacología , Emulsiones/uso terapéutico , Hígado/metabolismo , Modelos Biológicos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Fosfolípidos/administración & dosificación , Fosfolípidos/farmacología , Ratas , Aceite de Soja/administración & dosificación , Aceite de Soja/farmacología , Triglicéridos/administración & dosificación , Triglicéridos/farmacología
20.
Cardiovasc Toxicol ; 15(2): 127-39, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25134468

RESUMEN

Cardiovascular (CV) toxicity is a leading cause of drug attrition and withdrawal. Introducing in vitro assays with higher throughput should permit earlier CV hazard identification and enable medicinal chemists to design-out liabilities. Heretofore, development of in vitro CV assays has been limited by the challenge of replicating integrated cardiovascular physiology while achieving the throughput and consistency required for screening. These challenges appear to be met with a combination of human stem cell-derived cardiomyocytes (CM) which beat spontaneously and monitoring the response with technology that can assess drug-induced changes in voltage dependent contraction such as cellular impedance which has been validated with excellent predictivity for drug-induced arrhythmia and contractility. Here, we review advances in cardiomyocyte impedance with emphasis on stem cell-derived cardiomyocyte models for toxicity screening. Key perspectives include: the electrical principles of impedance technology, impedance detection of cardiomyocyte beating, beat parameter selection/analysis, validation in toxicity and drug discovery, and future directions. As a conclusion, an in vitro screening cascade is proffered using the downstream, inclusive detection of CM impedance assays as a primary screen followed by complementary CM assays chosen to enable mechanism-appropriate follow-up. The combined approach will enhance testing for CV liabilities prior to traditional in vivo models.


Asunto(s)
Cardiotoxinas/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Cardiotoxicidad/diagnóstico , Cardiotoxicidad/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Impedancia Eléctrica , Humanos , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Células Madre/patología , Células Madre/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA