Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
Más filtros

Tipo del documento
Publication year range
1.
Circ Res ; 126(8): 947-964, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32091972

RESUMEN

RATIONALE: Drug-induced proarrhythmia is so tightly associated with prolongation of the QT interval that QT prolongation is an accepted surrogate marker for arrhythmia. But QT interval is too sensitive a marker and not selective, resulting in many useful drugs eliminated in drug discovery. OBJECTIVE: To predict the impact of a drug from the drug chemistry on the cardiac rhythm. METHODS AND RESULTS: In a new linkage, we connected atomistic scale information to protein, cell, and tissue scales by predicting drug-binding affinities and rates from simulation of ion channel and drug structure interactions and then used these values to model drug effects on the hERG channel. Model components were integrated into predictive models at the cell and tissue scales to expose fundamental arrhythmia vulnerability mechanisms and complex interactions underlying emergent behaviors. Human clinical data were used for model framework validation and showed excellent agreement, demonstrating feasibility of a new approach for cardiotoxicity prediction. CONCLUSIONS: We present a multiscale model framework to predict electrotoxicity in the heart from the atom to the rhythm. Novel mechanistic insights emerged at all scales of the system, from the specific nature of proarrhythmic drug interaction with the hERG channel, to the fundamental cellular and tissue-level arrhythmia mechanisms. Applications of machine learning indicate necessary and sufficient parameters that predict arrhythmia vulnerability. We expect that the model framework may be expanded to make an impact in drug discovery, drug safety screening for a variety of compounds and targets, and in a variety of regulatory processes.


Asunto(s)
Antiarrítmicos/química , Arritmias Cardíacas/tratamiento farmacológico , Cardiotoxinas/química , Simulación por Computador , Descubrimiento de Drogas/métodos , Canal de Potasio ERG1/química , Antiarrítmicos/metabolismo , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/metabolismo , Cardiotoxicidad/metabolismo , Cardiotoxicidad/prevención & control , Cardiotoxinas/efectos adversos , Cardiotoxinas/metabolismo , Descubrimiento de Drogas/tendencias , Canal de Potasio ERG1/metabolismo , Femenino , Humanos , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/metabolismo , Aprendizaje Automático , Masculino , Moxifloxacino/química , Moxifloxacino/metabolismo , Moxifloxacino/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Fenetilaminas/química , Fenetilaminas/metabolismo , Fenetilaminas/uso terapéutico , Estructura Secundaria de Proteína , Sulfonamidas/química , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/metabolismo , Inhibidores de Topoisomerasa II/uso terapéutico
2.
Mol Cell Biochem ; 476(8): 3099-3109, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33835331

RESUMEN

While anthracyclines (ACs) are a class of chemotherapeutic agents that have improved the prognosis of many women with breast cancer, it is one of the most cardiotoxic agents used to treat cancer. Despite their reported dose-dependent cardiotoxicity, AC-based chemotherapy has become the mainstay of breast cancer therapy due to its efficacy. Elucidating the mechanisms of anthracycline-mediated cardiotoxicity and associated therapeutic interventions continue to be the main focus in the field of cardio-oncology. Herein, we summarized the current literature surrounding the mechanisms of anthracycline-induced cardiotoxicity, including the role of topoisomerase II inhibition, generation of reactive oxygen species, and elevations in free radicals. Furthermore, this review highlights the molecular mechanisms of potential cardioprotective interventions in this setting. The benefits of pharmaceuticals, including dexrazoxane, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, beta-blockers, statins, and antioxidants in this setting, are reviewed. Finally, the mechanisms of emerging preventative interventions within this patient population including nutraceuticals and aerobic exercise are explored.


Asunto(s)
Antraciclinas/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Cardiotónicos/uso terapéutico , Cardiotoxicidad/prevención & control , Animales , Neoplasias de la Mama/patología , Cardiotoxicidad/etiología , Cardiotoxicidad/patología , Cardiotoxinas/efectos adversos , Femenino , Humanos , Pronóstico , Factores de Riesgo
3.
Pediatr Hematol Oncol ; 38(5): 497-503, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33715575

RESUMEN

BACKGROUND: Anthracycline chemotherapy is used to treat a variety of cancers. However, late cardiac effects of anthracycline chemotherapy, such as subclinical left ventricular dilatation and/or dysfunction, have been observed in more than half of long-term survivors of childhood cancers. A major risk factor for anthracycline cardiotoxicity is intrauterine growth restriction (IUGR). We assessed the significance of IUGR as an important risk factor for late cardiotoxic effects of anthracycline therapy in asymptomatic long-term survivors of childhood cancers. MATERIALS AND METHODS: The study included 61 survivors of childhood cancers. Cardiac functions were prospectively studied using both conventional and non-conventional echocardiographic methods (two-dimensional speckle tracking echocardiography) after completion of the treatment. The patients were divided into two groups based on their birth weights: Group 1 (patients with IUGR) and Group 2 (patients with normal birth weight). RESULTS: Conventional echocardiography revealed a similar and normal range of left ventricle systolic and diastolic functions in both groups. However, global longitudinal and circumferential strain values demonstrated subclinical left ventricular systolic dysfunction in both groups as compared with normal reference strain values. Furthermore, Group 1 patients had significantly lower global longitudinal and circumferential strain and strain rate values than those in Group 2 patients. CONCLUSION: Asymptomatic long-term survivors of childhood cancers with a history of IUGR may have an increased risk of anthracycline cardiotoxicity due to the low content of mitochondrial DNA (mtDNA). IUGR is a risk factor for late anthracycline cardiotoxicity.


Asunto(s)
Antraciclinas/efectos adversos , Antineoplásicos/efectos adversos , Cardiotoxicidad/etiología , Retardo del Crecimiento Fetal , Neoplasias/tratamiento farmacológico , Adolescente , Adulto , Peso al Nacer , Supervivientes de Cáncer , Cardiotoxinas/efectos adversos , Niño , Estudios Transversales , Retardo del Crecimiento Fetal/etiología , Humanos , Estudios Prospectivos , Factores de Riesgo , Adulto Joven
4.
Toxicol Appl Pharmacol ; 406: 115213, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32877659

RESUMEN

The limited availability of human heart tissue and its complex cell composition are major limiting factors for the reliable testing of drug efficacy and toxicity. Recently, we developed functional human and pig heart slice biomimetic culture systems that preserve the viability and functionality of 300 µm heart slices for up to 6 days. Here, we tested the reliability of this culture system for testing the cardiotoxicity of anti-cancer drugs. We tested three anti-cancer drugs (doxorubicin, trastuzumab, and sunitinib) with known different mechanisms of cardiotoxicity at three concentrations and assessed the effect of these drugs on heart slice viability, structure, function and gene expression. Slices incubated with any of these drugs for 48 h showed diminished in viability as well as loss of cardiomyocyte structure and function. Mechanistically, RNA sequencing of doxorubicin-treated tissues demonstrated a significant downregulation of cardiac genes and upregulation of oxidative stress responses. Trastuzumab treatment downregulated cardiac muscle contraction-related genes consistent with its clinically known effect on cardiomyocytes. Interestingly, sunitinib treatment resulted in significant downregulation of angiogenesis-related genes, in line with its mechanism of action. Similar to hiPS-derived-cardiomyocytes, heart slices recapitulated the expected toxicity of doxorubicin and trastuzumab, however, slices were superior in detecting sunitinib cardiotoxicity and mechanism in the clinically relevant concentration range of 0.1-1 µM. These results indicate that heart slice culture models have the potential to become a reliable platform for testing and elucidating mechanisms of drug cardiotoxicity.


Asunto(s)
Cardiotoxicidad , Cardiotoxinas/efectos adversos , Corazón/efectos de los fármacos , Modelos Biológicos , Técnicas de Cultivo de Tejidos , Adulto , Anciano , Animales , Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Doxorrubicina/efectos adversos , Femenino , Corazón/fisiología , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Persona de Mediana Edad , Porcinos , Trastuzumab/efectos adversos
5.
J Muscle Res Cell Motil ; 41(4): 375-387, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32621158

RESUMEN

To improve muscle healing upon injury, it is of importance to understand the interplay of key signaling pathways during muscle regeneration. To study this, mice were injected with cardiotoxin (CTX) or PBS in the Tibialis Anterior muscle and were sacrificed 2, 5 and 12 days upon injection. The time points represent different phases of the regeneration process, i.e. destruction, repair and remodeling, respectively. Two days upon CTX-injection, p-mTORC1 signaling and stress markers such as BiP and p-ERK1/2 were upregulated. Phospho-ERK1/2 and p-mTORC1 peaked at d5, while BiP expression decreased towards PBS levels. Phospho-FOXO decreased 2 and 5 days following CTX-injection, indicative of an increase in catabolic signaling. Furthermore, CTX-injection induced a shift in the fiber type composition, characterized by an initial loss in type IIa fibers at d2 and at d5. At d5, new type IIb fibers appeared, whereas type IIa fibers were recovered at d12. To conclude, CTX-injection severely affected key modulators of muscle metabolism and histology. These data provide useful information for the development of strategies that aim to improve muscle molecular signaling and thereby recovery.


Asunto(s)
Cardiotoxinas/efectos adversos , Fibras Musculares Esqueléticas/metabolismo , Enfermedades Musculares/inducido químicamente , Animales , Masculino , Ratones , Transducción de Señal
6.
J Oncol Pharm Pract ; 26(2): 434-444, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31594518

RESUMEN

BACKGROUND: Doxorubicin is one of the most commonly prescribed and time-tested anticancer drugs. Although being considered as a first line drug in different types of cancers, the two main obstacles to doxorubicin therapy are drug-induced cardiotoxicity and drug resistance. METHOD: The study utilizes systemic reviews on publications of previous studies obtained from scholarly journal databases including PubMed, Medline, Ebsco Host, Google Scholar, and Cochrane. The study utilizes secondary information obtained from health organizations using filters and keywords to sustain information relevancy. The study utilizes information retrieved from studies captured in the peer-reviewed journals on "doxorubicin-induced cardiotoxicity" and "doxorubicin resistance." DISCUSSION AND RESULTS: The exact mechanisms of cardiotoxicity are not known; various hypotheses are studied. Doxorubicin can lead to free radical generation in various ways. The commonly proposed underlying mechanisms promoting doxorubicin resistance are the expression of multidrug resistance proteins as well as other causes. CONCLUSION: In this review, we have described the major obstacles to doxorubicin therapy, doxorubicin-induced cardiotoxicity as well as the mechanisms of cancer drug resistance and in following the treatment failures.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiotoxinas/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Doxorrubicina/efectos adversos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/uso terapéutico , Cardiotoxicidad , Cardiotoxinas/uso terapéutico , Enfermedades Cardiovasculares/diagnóstico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Neoplasias/diagnóstico
7.
Eur Heart J ; 40(22): 1764-1770, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29377985

RESUMEN

Cardiotoxic effects from cancer therapy are a major cause of morbidity during cancer treatment. Unexpected toxicity can occur during treatment and/or after completion of therapy, into the time of cancer survivorship. While older drugs such as anthracyclines have well-known cardiotoxic effects, newer drugs such as tyrosine kinase inhibitors, proteasome inhibitors, and immunotherapies also can cause diverse cardiovascular and metabolic complications. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are increasingly being used as instruments for disease modelling, drug discovery, and mechanistic toxicity studies. Promising results with hiPSC-CM chemotherapy studies are raising hopes for improving cancer therapies through personalized medicine and safer drug development. Here, we review the cardiotoxicity profiles of common chemotherapeutic agents as well as efforts to model them in vitro using hiPSC-CMs.


Asunto(s)
Antineoplásicos/efectos adversos , Cardiomiopatías/inducido químicamente , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas , Modelos Biológicos , Animales , Antineoplásicos/uso terapéutico , Cardiotoxicidad , Cardiotoxinas/efectos adversos , Genómica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Neoplasias/tratamiento farmacológico , Medicina de Precisión
8.
Eur Heart J ; 40(48): 3913-3920, 2019 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-31318428

RESUMEN

AIMS: Develop a score to predict the risk of major adverse cardiovascular events (MACE) after early stage breast cancer (EBC) to facilitate personalized decision-making about potentially cardiotoxic treatments and interventions to reduce cardiovascular risk. METHODS AND RESULTS: Using administrative databases, we assembled a cohort of women diagnosed with EBC in Ontario between 2003 and 2014, with follow-up through 2015. Two-thirds of the cohort were used for risk score derivation; the remainder were reserved for its validation. The outcome was a composite of hospitalizations for acute myocardial infarction, unstable angina, transient ischaemic attack, stroke, peripheral vascular disease, heart failure (HF), or cardiovascular death. We developed the score by regressing MACE incidence against candidate predictors in the derivation sample using a Fine-Gray model. Discrimination was assessed in the validation sample using Wolber's c-index for prognostic models with competing risks, while calibration was assessed by comparing predicted and observed MACE incidence. The risk score was derived in 60 294 women and validated in 29 810 women. Age, hypertension, diabetes, ischaemic heart disease, atrial fibrillation, HF, cerebrovascular disease, peripheral vascular disease, chronic obstructive pulmonary disease, and chronic kidney disease were significantly associated with MACE incidence and incorporated into the score. Ten-year MACE incidence was >40-fold higher for patients in the highest score decile compared to the lowest. The c-index was 81.9% (95% confidence interval 80.9-82.9%) at 5 years and 79.8% (78.8-80.8%) at 10 years in the validation cohort, with good agreement between predicted and observed MACE incidence. CONCLUSION: Cardiovascular prognosis after EBC can be estimated using patients' pre-treatment characteristics.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/patología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/prevención & control , Anciano , Cardiotoxinas/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Estudios de Casos y Controles , Trastornos Cerebrovasculares/epidemiología , Toma de Decisiones Clínicas , Estudios de Cohortes , Muerte , Diabetes Mellitus/epidemiología , Femenino , Hospitalización , Humanos , Incidencia , Ataque Isquémico Transitorio/epidemiología , Persona de Mediana Edad , Estadificación de Neoplasias , Ontario/epidemiología , Enfermedades Vasculares Periféricas/epidemiología , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Insuficiencia Renal Crónica/epidemiología , Medición de Riesgo , Accidente Cerebrovascular/epidemiología
9.
Int J Mol Sci ; 21(3)2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32023816

RESUMEN

Duchenne muscular dystrophy (DMD) is a genetic disease characterized by muscle wasting and chronic inflammation, leading to impaired satellite cells (SCs) function and exhaustion of their regenerative capacity. We previously showed that lack of PKCθ in mdx mice, a mouse model of DMD, reduces muscle wasting and inflammation, and improves muscle regeneration and performance at early stages of the disease. In this study, we show that muscle regeneration is boosted, and fibrosis reduced in mdxθ-/- mice, even at advanced stages of the disease. This phenotype was associated with a higher number of Pax7 positive cells in mdxθ-/- muscle compared with mdx muscle, during the progression of the disease. Moreover, the expression level of Pax7 and Notch1, the pivotal regulators of SCs self-renewal, were upregulated in SCs isolated from mdxθ-/- muscle compared with mdx derived SCs. Likewise, the expression of the Notch ligands Delta1 and Jagged1 was higher in mdxθ-/- muscle compared with mdx. The expression level of Delta1 and Jagged1 was also higher in PKCθ-/- muscle compared with WT muscle following acute injury. In addition, lack of PKCθ prolonged the survival and sustained the differentiation of transplanted myogenic progenitors. Overall, our results suggest that lack of PKCθ promotes muscle repair in dystrophic mice, supporting stem cells survival and maintenance through increased Delta-Notch signaling.


Asunto(s)
Cardiotoxinas/efectos adversos , Músculo Esquelético/lesiones , Distrofia Muscular de Duchenne/genética , Proteína Quinasa C-theta/genética , Células Madre/citología , Animales , Diferenciación Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Factor de Transcripción PAX7/metabolismo , Receptor Notch1/metabolismo , Regeneración , Transducción de Señal , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Células Madre/metabolismo
10.
Zhonghua Zhong Liu Za Zhi ; 42(10): 838-842, 2020 Oct 23.
Artículo en Zh | MEDLINE | ID: mdl-33113624

RESUMEN

Cardiovascular toxicity of cancer patients in antineoplastic therapy is gradually paid widespread attention. Although many high-risk factors of cardiovascular toxicity associated with chemotherapy, targeted therapy or immunotherapy have been identified, it is still difficult to establish accurate risk prediction model. Traditional risk prediction model cannot adequately explain the differences in cardiovascular toxicity susceptibility among patients, makes it difficult to accurately screen high-risk groups, early diagnose and prevent cardiovascular toxicity. Finding susceptible genes of cardiovascular toxicity associated with antineoplastic therapy and incorporating single-nucleotide polymorphisms into risk prediction model can significantly improve the identification of high-risk population of cardiovascular toxicity.


Asunto(s)
Antineoplásicos , Sistema Cardiovascular/efectos de los fármacos , Neoplasias/genética , Antineoplásicos/efectos adversos , Cardiotoxinas/efectos adversos , Humanos , Modelos Teóricos , Neoplasias/tratamiento farmacológico , Polimorfismo de Nucleótido Simple , Medición de Riesgo
11.
Mol Cell Biochem ; 455(1-2): 41-59, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30390173

RESUMEN

The aim of this study was to examine the potential association between the expression of Hsp70 protein and heart failure and to investigate the possible protective effect of Hsp70 against the doxorubicin-induced toxicity. Initially, at clinical level, the expression levels of the inducible Hsp70 were quantified in serum from patients with heart failure. Our results showed that in heart failure, Hsp70 concentration appeared to be increased in blood sera of patients compared to that of healthy individuals. The enhanced expression of Hsp70 in serum of patients with heart failure seemed to be associated with various features, such as gender, age and the type of heart failure, but not with its etiology. Next, in our study at cellular level, we used primary cell cultures isolated from embryos of Hsp70-transgenic mice (Tg/Tg) overexpressing human HSP70 and wild-type mice (F1/F1). After exposure to a wide range of doxorubicin concentrations and incubation times, the dose- and time-dependent toxicity of the drug, which appeared to be reduced in Tg/Tg cells, was demonstrated. In addition, doxorubicin administration appeared to result in a dose- and time-dependent decrease in the activity of two of the major endogenous antioxidant enzymes (SOD and GPx). The increased activity of these enzymes in Tg/Tg cells compared to the control F1/F1 cells was obvious, suggesting that the presence of Hsp70 confers enhanced tolerance against DOX-induced oxidative stress. Overall, it has been indicated that Hsp70 protein exerts a very important protective action and renders cells more resistant to the harmful effects of doxorubicin.


Asunto(s)
Cardiotoxinas/efectos adversos , Doxorrubicina/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/sangre , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/inducido químicamente , Animales , Cardiotoxinas/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Proteínas HSP70 de Choque Térmico/genética , Insuficiencia Cardíaca/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética
12.
J Biol Regul Homeost Agents ; 33(5): 1425-1436, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31576730

RESUMEN

Aconitine (ACO), the main active component in Aconitum carmichaelii Debeaux (family: Ranunculaceae), has high cardiotoxicity, however the mechanisms of this effect remain unclear. Paeoniflorin (PF), the main chemical ingredient in herbaceous peony, can protect the heart from damage through antioxidant, vasodilatory and other effects. In this study, we focused on the mechanism by which PF reduces ACO cardiotoxicity. We selected H9c2 cells as the experimental model. MTT assay, Western blot analysis and real-time PCR were used to measure cell proliferation, apoptosis, ion channels and oxidative stress. Cell proliferation was significantly increased, the Bcl-2/Bax ratio and p53 level were upregulated, and Caspase-3 was slightly reduced in the ACO+PF group compared with the ACO group. SCN5A mRNA expression was significantly increased in the ACO+PF group compared with the ACO group, while RyR2 and Cx43 mRNA expression was decreased. Compared with the ACO group, the ACO+PF group showed marked decreases in extracellular lactate dehydrogenase (LDH) and intracellular malondialdehyde (MDA), while there was no difference in intracellular superoxide dismutase (SOD). The above data demonstrate that the cardiotoxicity of ACO in H9c2 cells was significantly decreased by PF.


Asunto(s)
Aconitina/efectos adversos , Cardiotoxinas/efectos adversos , Glucósidos/farmacología , Monoterpenos/farmacología , Mioblastos/efectos de los fármacos , Animales , Apoptosis , Línea Celular , Estrés Oxidativo , Ratas
13.
Eur Heart J ; 39(43): 3896-3903, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29635274

RESUMEN

Aims: Breast cancer survival has improved throughout the last decades, but treatment-induced cardiotoxicity remains a major concern. This study aimed to investigate competing causes of death and prognostic factors within a large cohort of breast cancer patients and to describe the heart-specific mortality in relation to the general population. Methods and results: In this registry-based cohort study, women diagnosed with breast cancer between 2000 and 2011, who were treated with radiotherapy or chemotherapy and followed until 2014, were identified from the Surveillance, Epidemiology, and End Results-18 (SEER-18) database. Cumulative mortality functions were computed. To investigate heart-specific mortality relative to the general population, long-term (≥10 years) standardized mortality ratios (SMRs) were calculated. Prognostic factors for heart-specific mortality were assessed by calculating cause-specific hazard ratios (HRcs) with corresponding 95% confidence intervals using the Cox proportional hazards regression. Subgroup analysis on intermediate-term mortality according to molecular subtypes, for which information was available since 2010, was performed. In total, 347 476 breast cancer patients were eligible to be included in the study. Among all possible competing causes of death, breast cancer accounted for the highest cumulative mortality. Compared with the general population, heart-specific mortality of breast cancer patients treated with radiotherapy or chemotherapy was lower [SMRoverall 0.84 (0.79-0.90)]. In subgroup analysis, human epidermal growth factor receptor 2 (HER2)-positive subtype was not associated with increased heart-specific mortality relative to HER2-negative patients [HRcs 0.96 (0.70-1.32)]. Conclusion: Heart-specific mortality among breast cancer survivors is not increased compared with the general population. Human epidermal growth factor receptor 2-positive patients do not have increased heart-specific mortality compared to HER2-negative patients.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias de la Mama , Cardiotoxinas/efectos adversos , Enfermedades Cardiovasculares , Radioterapia/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/mortalidad , Femenino , Humanos , Persona de Mediana Edad , Sistema de Registros , Estudios Retrospectivos
14.
Biochem Biophys Res Commun ; 503(4): 3038-3043, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30170730

RESUMEN

Abnormally expressed long noncoding RNAs (lncRNAs) has been recognized as one of the key source in cardiac diseases. However, the role of lncRNA in doxorubicin (DOX)-induced cardiotoxicity remains largely unknown. In previous studies, we have screened some aberrantly expressed lncRNAs from an animal model for DOX-induced cardiotoxicity, and LINC00339 is one of the highly expressed lncRNA. In this study, we validated and further explored its regulatory mechanisms using in vitro model systems. Primary cultured myocardial cell (PC) and H9C2 cell line were treated with different concentrations of DOX and the expression of LINC00339 were markedly up-regulated. However, knockdown of endogenous LINC00339 by its siRNA improved cells proliferation activity and reduced cardiomyocyte apoptosis. Further experiments showed the opposite trend of expression between LINC00339 and miR-484. Bioinformatics analysis and luciferase reporter assay indicated that LINC00339 directly binds to miR-484. Moreover, miR-484 inhibitor abrogated the collagen synthesis inhibition induced by LINC00339. These findings reveal a novel function of the LINC00339/miR-484 axis in DOX-induced cardiotoxicity.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiotoxinas/efectos adversos , Doxorrubicina/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Miocitos Cardíacos/efectos de los fármacos , ARN Largo no Codificante/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Células Cultivadas , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
15.
Int J Mol Sci ; 19(8)2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-30111743

RESUMEN

Chronic effects of a combination antiretroviral therapy (cART = tenofovir/emtricitatine + atazanavir/ritonavir) on systemic and cardiac oxidative stress/injury in HIV-1 transgenic (Tg) rats and protection by Mg-supplementation were assessed. cART (low doses) elicited no significant effects in normal rats, but induced time-dependent oxidative/nitrosative stresses: 2.64-fold increased plasma 8-isoprostane, 2.0-fold higher RBC oxidized glutathione (GSSG), 3.2-fold increased plasma 3-nitrotyrosine (NT), and 3-fold elevated basal neutrophil superoxide activity in Tg rats. Increased NT staining occurred within cART-treated HIV-Tg hearts, and significant decreases in cardiac systolic and diastolic contractile function occurred at 12 and 18 weeks. HIV-1 expression alone caused modest levels of oxidative stress and cardiac dysfunction. Significantly, cART caused up to 24% decreases in circulating Mg in HIV-1-Tg rats, associated with elevated renal NT staining, increased creatinine and urea levels, and elevated plasma substance P levels. Strikingly, Mg-supplementation (6-fold) suppressed all oxidative/nitrosative stress indices in the blood, heart and kidney and substantially attenuated contractile dysfunction (>75%) of cART-treated Tg rats. In conclusion, cART caused significant renal and cardiac oxidative/nitrosative stress/injury in Tg-rats, leading to renal Mg wasting and hypomagnesemia, triggering substance P-dependent neurogenic inflammation and cardiac dysfunction. These events were effectively attenuated by Mg-supplementation likely due to its substance P-suppressing and Mg's intrinsic anti-peroxidative/anti-calcium properties.


Asunto(s)
Antirretrovirales/efectos adversos , Corazón/efectos de los fármacos , Magnesio/uso terapéutico , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/prevención & control , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/uso terapéutico , Animales , Terapia Antirretroviral Altamente Activa/efectos adversos , Cardiotoxinas/efectos adversos , Expresión Génica , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , VIH-1/genética , Corazón/fisiopatología , Riñón/efectos de los fármacos , Riñón/fisiopatología , Masculino , Inflamación Neurogénica/fisiopatología , Activación Neutrófila/efectos de los fármacos , Estrés Nitrosativo/efectos de los fármacos , Ratas Endogámicas F344 , Ratas Transgénicas
16.
Circulation ; 133(13): 1272-89, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-27022039

RESUMEN

Since the late 1990s, there has been a steady decline in cancer-related mortality, in part related to the introduction of so-called targeted therapies. Intended to interfere with a specific molecular pathway, these therapies have, paradoxically, led to a number of effects off their intended cancer tissue or molecular targets. The latest examples are tyrosine kinase inhibitors targeting the Philadelphia Chromosome mutation product, which have been associated with progressive atherosclerosis and acute vascular events. In addition, agents designed to interfere with the vascular growth factor signaling pathway have vascular side effects ranging from hypertension to arterial events and cardiomyocyte toxicity. Interestingly, the risk of cardiotoxicity with drugs such as trastuzumab is predicted by preexisting cardiovascular risk factors and disease, posing the question of a vascular component to the pathophysiology. The effect on the coronary circulation has been the leading explanation for the cardiotoxicity of 5-fluorouracil and may be the underlying the mechanism of presentation of apical ballooning syndrome with various chemotherapeutic agents. Classical chemotherapeutic agents such as cisplatin, often used in combination with bleomycin and vinca alkaloids, can lead to vascular events including acute coronary thrombosis and may be associated with an increased long-term cardiovascular risk. This review is intended to provide an update on the evolving spectrum of vascular toxicities with cancer therapeutics, particularly as they pertain to clinical practice, and to the conceptualization of cardiovascular diseases, as well. Vascular toxicity with cancer therapy: the old and the new, an evolving avenue.


Asunto(s)
Antineoplásicos/efectos adversos , Cardiotoxinas/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Neoplasias/tratamiento farmacológico , Animales , Enfermedades Cardiovasculares/diagnóstico , Humanos , Neoplasias/diagnóstico
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(8): 1904-1911, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28069395

RESUMEN

Doxorubicin as anticancer agent can cause dose-dependent cardiotoxicity and heart failure in the long term. Rutin as a polyphenolic flavonoid has been illustrated to protect hearts from diverse cardiovascular diseases. Its function is known to be related to its antioxidant and antiinflammatory activity which may regulate multiple cellular signal pathways. However, the role of rutin on doxorubicin-induced cardiotoxicity has yet to be discovered. In this study, we explored the protective role of rutin on doxorubicin-induced heart failure and elucidated the potential mechanisms of protective effects of rutin against cardiomyocyte death. We analyzed cardiac tissues at the time point of 8weeks after doxorubicin treatment. The results by echocardiography, TUNEL staining, Masson's trichrome staining as well as Western blot analysis revealed that doxorubicin induced remarkable cardiac dysfunction and cardiotoxicity in mice hearts and cardiomyocytes, which were alleviated by rutin treatment. Western blot analysis indicated that the underlying mechanisms included inhibition excessive autophagy and apoptosis mediated by Akt activation. Collectively, our findings suggest that suppression of autophagy and apoptosis by administration of rutin could attenuate doxorubicin-induced cardiotoxicity, which enhances our knowledge to explore new drugs and strategies for combating this devastating side effect induced by doxorubicin. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Cardiotoxinas/efectos adversos , Doxorrubicina/efectos adversos , Rutina/farmacología , Animales , Cardiotoxinas/farmacología , Doxorrubicina/farmacología , Masculino , Ratones
18.
Cardiovasc Diabetol ; 16(1): 113, 2017 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-28903775

RESUMEN

BACKGROUND: In the Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus (SAVOR) trial in patients with type 2 diabetes mellitus (T2D) at high risk of cardiovascular (CV) disease, saxagliptin did not increase the risk for major CV adverse events. However, there was an unexpected imbalance in events of hospitalization for heart failure (hHF), one of six components of the secondary CV composite endpoint, with a greater number of events observed with saxagliptin. Here, we examined findings from nonclinical safety and clinical pharmacology studies of saxagliptin with the aim of identifying any potential signals of myocardial injury. METHODS: In vitro and in vivo (rat, dog, monkey) safety pharmacology and toxicology studies evaluating the potential effects of saxagliptin and its major active metabolite, 5-hydroxy saxagliptin, on the CV system are reviewed. In addition, results from saxagliptin clinical studies are discussed: one randomized, 2-period, double-blind, placebo-controlled single-ascending-dose study (up to 100 mg); one randomized, double-blind, placebo-controlled, sequential, multiple-ascending-high-dose study (up to 400 mg/day for 14 days); and one randomized, double-blind, 4-period, 4-treatment, cross-over thorough QTc study (up to 40 mg/day for 4 days) in healthy volunteers; as well as one randomized, placebo-controlled, sequential multiple-ascending-dose study in patients with T2D (up to 50 mg/day for 14 days). RESULTS: Neither saxagliptin nor 5-hydroxy saxagliptin affected ligand binding to receptors and ion channels (e.g. potassium channels) or action potential duration in in vitro studies. In animal toxicology studies, no changes in the cardiac conduction system, blood pressure, heart rate, contractility, heart weight, or heart histopathology were observed. In healthy participants and patients with T2D, there were no findings suggestive of myocyte injury or fluid overload. Serum chemistry abnormalities indicative of cardiac injury, nonspecific muscle damage, or fluid homeostasis changes were infrequent and balanced across treatment groups. There were no QTc changes associated with saxagliptin. No treatment-emergent adverse events suggestive of heart failure or myocardial damage were reported. CONCLUSIONS: The saxagliptin nonclinical and clinical pharmacology programs did not identify evidence of myocardial injury and/or CV harm that may have predicted or may explain the unexpected imbalance in the rate of hHF observed in SAVOR.


Asunto(s)
Adamantano/análogos & derivados , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipéptidos/uso terapéutico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Adamantano/efectos adversos , Adamantano/uso terapéutico , Animales , Cardiotoxinas/efectos adversos , Cardiotoxinas/uso terapéutico , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/epidemiología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Dipéptidos/efectos adversos , Inhibidores de la Dipeptidil-Peptidasa IV/efectos adversos , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos
19.
PLoS Comput Biol ; 12(11): e1005214, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27870850

RESUMEN

The clinical use of the anthracycline doxorubicin is limited by its cardiotoxicity which is associated with mitochondrial dysfunction. Redox cycling, mitochondrial DNA damage and electron transport chain inhibition have been identified as potential mechanisms of toxicity. However, the relative roles of each of these proposed mechanisms are still not fully understood. The purpose of this study is to identify which of these pathways independently or in combination are responsible for doxorubicin toxicity. A state of the art mathematical model of the mitochondria including the citric acid cycle, electron transport chain and ROS production and scavenging systems was extended by incorporating a novel representation for mitochondrial DNA damage and repair. In silico experiments were performed to quantify the contributions of each of the toxicity mechanisms to mitochondrial dysfunction during the acute and chronic stages of toxicity. Simulations predict that redox cycling has a minor role in doxorubicin cardiotoxicity. Electron transport chain inhibition is the main pathway for acute toxicity for supratherapeutic doses, being lethal at mitochondrial concentrations higher than 200µM. Direct mitochondrial DNA damage is the principal pathway of chronic cardiotoxicity for therapeutic doses, leading to a progressive and irreversible long term mitochondrial dysfunction.


Asunto(s)
Cardiotoxinas/efectos adversos , ADN Mitocondrial/genética , Doxorrubicina/efectos adversos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Modelos Biológicos , Enfermedad Aguda , Animales , Antibióticos Antineoplásicos/efectos adversos , Enfermedad Crónica , Ciclo del Ácido Cítrico/efectos de los fármacos , Simulación por Computador , Daño del ADN , Reparación del ADN/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteínas del Complejo de Cadena de Transporte de Electrón/efectos de los fármacos , Humanos , Mitocondrias Cardíacas/patología , Especies Reactivas de Oxígeno/metabolismo
20.
Pharm Res ; 34(9): 1849-1856, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28560697

RESUMEN

PURPOSE: MicroRNAs (miRs) are a group of small non-coding RNAs that regulate transcriptional or post-transcriptional gene expression. The aim of the present study was to investigate the role of miR -1, -21 and -145 and their targets in cardiotoxicity-induced by DOX and pegylated liposomal DOX. METHODS: BALB/c mice subjected to subcutaneous injection of C-26 tumor cells. Eight days after tumor inoculation, animals were divided into 6 groups: control, liposome, DOX (6 and 9 mg/kg) and PL-DOX (6 and 9 mg/kg). The formulations were administered one time per week for four weeks. 24 h after the last injection, mice were sacrificed; blood and heart samples were taken. Western blot analysis was done on protein extracts to investigate the expression of cardiac caspase-3, -8, Bax, Bcl2, Programmed cell death 4 (PDCD4) and BCL2/Adenovirus E1B 19 kDa Interacting Protein 3 (BNIP3). The expression levels of miR -1, -21 and -145 were also evaluated by quantitative real-time PCR. RESULTS: Mice treated with both DOX formulations showed a marked inhibition in tumor growth. Western blot analysis indicated that the expression level of cardiac caspase-3, caspase-8, Bax and BNIP3 were up-regulated due to DOX injection (9 mg/kg). Exposure of mice with DOX resulted in a significant increase in cardiac miR-1 and miR-21 expression level. PL-DOX treatment did not change the proteins and miRs expression. CONCLUSION: The results suggest that miR -1, -21 and -145 may involve in cardiotoxicity induced by DOX. Evaluation of miRs signaling pathways might be of potential value for toxicity assessment of new formulations. Graphical Abstract The cardiotoxic mechanism of doxorubicin (DOX) and pegylated liposomal DOX (PL-DOX).


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiotoxinas/efectos adversos , Neoplasias del Colon/tratamiento farmacológico , Doxorrubicina/análogos & derivados , Corazón/efectos de los fármacos , MicroARNs/genética , Polietilenglicoles/efectos adversos , Animales , Línea Celular Tumoral , Neoplasias del Colon/genética , Doxorrubicina/efectos adversos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda