Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 127: 4-14, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28336372

RESUMEN

Anthracycline cardiotoxicity remains a serious problem in paediatric and adult cancer survivors, and the advancement of cardio-oncology is a necessary step for an effective care of the patients that experience adverse cardiovascular effects. In this review, we discuss the multiple instruments used by clinicians that constitute the current strategies for primary and secondary prevention aiming at contrasting the onset of early and late doxorubicin-induced cardiotoxic events. The importance of early detection of cardiotoxicity and the following pharmacological therapy has been acknowledged with the emphasis put on impaired diastolic function, an increasingly recognized precocious sign of doxorubicin cardiotoxicity with an emerging scientific and clinical interest. We highlight the involvement of progenitor cells of cardiac and extra-cardiac origin as well as multiple cardiac cell types (fibroblasts and vasculature cells), focusing on molecular signals involved in cellular injury and response. Oxidative stress, DNA damage, senescence and cell death are established mechanisms driving anthracycline toxicity, but the comprehension of their relative weight on affecting specific cell type behaviour remains to be consolidated. The contribution of these crucial stressors and the emerging tools for preserving cell function are discussed.


Asunto(s)
Cardiotoxicidad/prevención & control , Doxorrubicina/efectos adversos , Biomarcadores Farmacológicos , Cardiotoxicidad/tratamiento farmacológico , Humanos , Modelos Biológicos
2.
Mediators Inflamm ; 2016: 3917471, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28090152

RESUMEN

Structural changes contribute to airway hyperresponsiveness and airflow obstruction in asthma. Emerging evidence points to the involvement of c-kit+ cells in lung homeostasis, although their potential role in asthma is unknown. Our aim was to isolate c-kit+ cells from normal mouse lungs and to test whether these cells can interfere with hallmarks of asthma in an animal model. Adult mouse GFP-tagged c-kit+ cells, intratracheally delivered in the ovalbumin-induced airway hyperresponsiveness, positively affected airway remodeling and improved airway function. In bronchoalveolar lavage fluid of cell-treated animals, a reduction in the number of inflammatory cells and in IL-4, IL-5, and IL-13 release, along with an increase of IL-10, was observed. In MSC-treated mice, the macrophage polarization to M2-like subset may explain, at least in part, the increment in the level of anti-inflammatory cytokine IL-10. After in vitro stimulation of c-kit+ cells with proinflammatory cytokines, the indoleamine 2,3-dioxygenase and TGFß were upregulated. These data, together with the increased apoptosis of inflammatory cells in vivo, indicate that c-kit+ cells downregulate immune response in asthma by influencing local environment, possibly by cell-to-cell contact combined to paracrine action. In conclusion, intratracheally administered c-kit+ cells reduce inflammation, positively modulate airway remodeling, and improve function. These data document previously unrecognized properties of c-kit+ cells, able to impede pathophysiological features of experimental airway hyperresponsiveness.


Asunto(s)
Hiperreactividad Bronquial/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Asma/inmunología , Lavado Broncoalveolar , Líquido del Lavado Bronquioalveolar/química , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/química , Homeostasis , Sistema Inmunológico , Inflamación , Interleucina-10/uso terapéutico , Pulmón/patología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos BALB C
3.
Mol Pharmacol ; 84(4): 603-14, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23913256

RESUMEN

It is known that glutamate (Glu), the major excitatory amino acid in the central nervous system, can be an essential source for cell energy metabolism. Here we investigated the role of the plasma membrane Na(+)/Ca(2+) exchanger (NCX) and the excitatory amino acid transporters (EAATs) in Glu uptake and recycling mechanisms leading to ATP synthesis. We used different cell lines, such as SH-SY5Y neuroblastoma, C6 glioma and H9c2 as neuronal, glial, and cardiac models, respectively. We first observed that Glu increased ATP production in SH-SY5Y and C6 cells. Pharmacological inhibition of either EAAT or NCX counteracted the Glu-induced ATP synthesis. Furthermore, Glu induced a plasma membrane depolarization and an intracellular Ca(2+) increase, and both responses were again abolished by EAAT and NCX blockers. In line with the hypothesis of a mutual interplay between the activities of EAAT and NCX, coimmunoprecipitation studies showed a physical interaction between them. We expanded our studies on EAAT/NCX interplay in the H9c2 cells. H9c2 expresses EAATs but lacks endogenous NCX1 expression. Glu failed to elicit any significant response in terms of ATP synthesis, cell depolarization, and Ca(2+) increase unless a functional NCX1 was introduced in H9c2 cells by stable transfection. Moreover, these responses were counteracted by EAAT and NCX blockers, as observed in SH-SY5Y and C6 cells. Collectively, these data suggest that plasma membrane EAAT and NCX are both involved in Glu-induced ATP synthesis, with NCX playing a pivotal role.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Membrana Celular/metabolismo , Transportador 1 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/farmacología , Intercambiador de Sodio-Calcio/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Ratas
4.
Basic Res Cardiol ; 108(2): 334, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23411815

RESUMEN

The increasing population of cancer survivors faces considerable morbidity and mortality due to late effects of the antineoplastic therapy. Cardiotoxicity is a major limiting factor of therapy with doxorubicin (DOXO), the most effective anthracycline, and is characterized by a dilated cardiomyopathy that can develop even years after treatment. Studies in animals have proposed the cardiac progenitor cells (CPCs) as the cellular target responsible for DOXO-induced cardiomyopathy but the relevance of these observations to clinical settings is unknown. In this study, the analysis of the DOXO-induced cardiomyopathic human hearts showed that the majority of human CPCs (hCPCs) was senescent. In isolated hCPCs, DOXO triggered DNA damage response leading to apoptosis early after exposure, and telomere shortening and senescence at later time interval. Functional properties of hCPCs, such as migration and differentiation, were also negatively affected. Importantly, the differentiated progeny of DOXO-treated hCPCs prematurely expressed the senescence marker p16(INK4a). In conclusion, DOXO exposure severely affects the population of hCPCs and permanently impairs their function. Premature senescence of hCPCs and their progeny can be responsible for the decline in the regenerative capacity of the heart and may represent the cellular basis of DOXO-induced cardiomyopathy in humans.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiomiopatía Dilatada/inducido químicamente , Senescencia Celular/efectos de los fármacos , Doxorrubicina/efectos adversos , Mioblastos Cardíacos/efectos de los fármacos , Células Madre/efectos de los fármacos , Adulto , Antibióticos Antineoplásicos/uso terapéutico , Western Blotting , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Muerte Celular/efectos de los fármacos , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de los fármacos , Doxorrubicina/uso terapéutico , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/metabolismo , Homeostasis del Telómero , beta-Galactosidasa/metabolismo
5.
Brain Sci ; 13(8)2023 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-37626577

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays a key role in brain development, contributing to neuronal survival and neuroplasticity. Previous works have found that BDNF is involved in several neurological or psychiatric diseases. In this review, we aimed to collect all available data on BDNF and bipolar disorder (BD) and assess if BDNF could be considered a biomarker for BD. We searched the most relevant medical databases and included studies reporting original data on BDNF circulating levels or Val66Met polymorphism. Only articles including a direct comparison with healthy controls (HC) and patients diagnosed with BD according to international classification systems were included. Of the 2430 identified articles, 29 were included in the present review. Results of the present review show a reduction in BDNF circulating levels during acute phases of BD compared to HC, which increase after effective therapy of the disorders. The Val66Met polymorphism was related to features usually associated with worse outcomes. High heterogeneity has been observed regarding sample size, clinical differences of included patients, and data analysis approaches, reducing comparisons among studies. Although more studies are needed, BDNF seems to be a promising biomarker for BD.

6.
Riv Psichiatr ; 58(6): 293-301, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38032033

RESUMEN

INTRODUCTION AND AIMS: Bipolar disorder (BD) is a severe and recurring mental illness associated with a significant personal and social burden. It has been recently hypothesized that increased levels of pro-inflammatory cytokines and cortisol, which is also associated with a reduced expression of the brain-derived neurotrophic factor (BDNF), may influence affective recurrences in BD. Our study aims to: 1) assess changes in the levels of peripheral cytokines, BDNF and salivary cortisol during acute and euthymic phases of bipolar disorder, compared to that of a sample oh healthy controls; 2) evaluate whether these changes represent a biosignature for the different phases of the illness. MATERIALS AND METHODS: Patients aged 18-65 years old, with a diagnosis of BD I or II types, will be enrolled during an acute episode, according to DSM-5 criteria, together with age- and gender-matched healthy controls. Blood and salivary samples will be collected at baseline and after 3 and 6 months. Validated assessment instruments will be administered to all participants for the evaluation of symptom severity, global functioning, suicidal risk, stress levels and physical comorbidities. EXPECTED RESULTS: We expect changes in inflammatory and neuroendocrine indices to be predictive of the onset of an acute phase of bipolar disorder and that overall levels of cytokines, cortisol and BDNF are overall significantly different between BD patients and healthy controls. CONCLUSIONS: The longitudinal design of the study will allow to assess whether the presence of acute affective symptoms in BD patients correlates with significantly higher levels of cytokines and salivary cortisol and with reduced BDNF levels compared to euthymic phases. Moreover, the comparison with healthy control subjects will allow to understand if inflammatory mediators as well as the hypothalamic-pituitary-adrenal (HPA) axis are chronically elevated in BD patients and are independent from mood swings.


Asunto(s)
Trastorno Bipolar , Adolescente , Adulto , Anciano , Humanos , Persona de Mediana Edad , Adulto Joven , Trastorno Bipolar/complicaciones , Factor Neurotrófico Derivado del Encéfalo , Citocinas , Hidrocortisona , Trastornos de la Personalidad , Masculino , Femenino , Ensayos Clínicos Controlados como Asunto
7.
J Cell Mol Med ; 16(4): 936-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21790971

RESUMEN

Although low-energy extracorporeal cardiac shock wave (ECSW) therapy represents an attractive non-invasive treatment option for ischaemic heart disease, the precise mechanisms of its action and influence on the cardiac tissue remain obscure. The goal of this study was to evaluate the effects of SW application on cardiac function and structure. Four-month-old Fisher 344 rats were subjected to ECSW therapy. Echocardiographic measurements of cardiac function were performed at baseline and at 1 and 3 months after treatment. Signs of inflammation, apoptosis and fibrosis were evaluated by immunohistochemistry in the control and treated hearts. ECSW application did not provoke arrhythmia or increase the troponin-I level. At all time points, the left ventricular ejection fraction and fractional shortening remained stable. Histological analysis revealed neither differences in the extracellular matrix collagen content nor the presence of fibrosis; similarly, there were no signs of inflammation. Moreover, a population of cardiac cells that responded eagerly to ECSW application in the adult heart was identified; c-kit-positive, Ki67-positive, orthochromatic cells, corresponding to cardiac primitive cells, were 2.65-fold more numerous in the treated myocardium. In conclusion, non-invasive ECSW therapy is a safe and effective way of activating cardiac stem cells and myocardial regeneration. Because many factors influence cellular turnover in the ischaemic myocardium during the course of ischaemic heart disease, cardiac remodelling, and heart failure progression, studies to identify the optimal treatment time are warranted.


Asunto(s)
Isquemia Miocárdica/terapia , Animales , Masculino , Isquemia Miocárdica/fisiopatología , Ratas , Ratas Endogámicas F344 , Regeneración
8.
Circulation ; 121(2): 276-92, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20038740

RESUMEN

BACKGROUND: Anthracyclines are the most effective drugs available in the treatment of neoplastic diseases; however, they have profound consequences on the structure and function of the heart, which over time cause a cardiomyopathy that leads to congestive heart failure. METHODS AND RESULTS: Administration of doxorubicin in rats led to a dilated myopathy, heart failure, and death. To test whether the effects of doxorubicin on cardiac anatomy and function were mediated by alterations in cardiac progenitor cells (CPCs), these cells were exposed to the anthracycline, which increased the formation of reactive oxygen species and caused increases in DNA damage, expression of p53, telomere attrition, and apoptosis. Additionally, doxorubicin resulted in cell-cycle arrest at the G2/M transition, which led to a significant decrease in CPC growth. Doxorubicin elicited multiple molecular adaptations; the massive apoptotic death that occurred in CPCs in the presence of anthracycline imposed on the surviving CPC pool the activation of several pathways aimed at preservation of the primitive state, cell division, lineage differentiation, and repair of damaged DNA. To establish whether delivery of syngeneic progenitor cells opposed the progression of doxorubicin cardiotoxicity, enhanced green fluorescent protein-labeled CPCs were injected in the failing myocardium; this treatment promoted regeneration of cardiomyocytes and vascular structures, which improved ventricular performance and rate of animal survival. CONCLUSIONS: Our results raise the possibility that autologous CPCs can be obtained before antineoplastic drugs are given to cancer patients and subsequently administered to individuals who are particularly sensitive to the cardiotoxicity of these agents for prevention or management of heart failure.


Asunto(s)
Antraciclinas/efectos adversos , Cardiomiopatías/inducido químicamente , Cardiomiopatías/terapia , Regeneración , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Animales , Cardiomiopatías/patología , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/terapia , Recuento de Células , Doxorrubicina/efectos adversos , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Humanos , Miocitos Cardíacos , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Ratas , Células Madre/fisiología
9.
Sci Rep ; 10(1): 12250, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32704131

RESUMEN

Cardiotoxicity remains a serious problem in anthracycline-treated oncologic patients. Therapeutic modulation of microRNA expression is emerging as a cardioprotective approach in several cardiovascular pathologies. MiR-34a increased in animals and patients exposed to anthracyclines and is involved in cardiac repair. In our previous study, we demonstrated beneficial effects of miR-34a silencing in rat cardiac cells exposed to doxorubicin (DOXO). The aim of the present work is to evaluate the potential cardioprotective properties of a specific antimiR-34a (Ant34a) in an experimental model of DOXO-induced cardiotoxicity. Results indicate that in our model systemic administration of Ant34a completely silences miR-34a myocardial expression and importantly attenuates DOXO-induced cardiac dysfunction. Ant34a systemic delivery in DOXO-treated rats triggers an upregulation of prosurvival miR-34a targets Bcl-2 and SIRT1 that mediate a reduction of DOXO-induced cardiac damage represented by myocardial apoptosis, senescence, fibrosis and inflammation. These findings suggest that miR-34a therapeutic inhibition may have clinical relevance to attenuate DOXO-induced toxicity in the heart of oncologic patients.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Doxorrubicina/efectos adversos , Silenciador del Gen , Predisposición Genética a la Enfermedad , MicroARNs/genética , Animales , Antibióticos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Cardiotoxicidad/diagnóstico , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Modelos Animales de Enfermedad , Doxorrubicina/uso terapéutico , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Genes bcl-2 , Modelos Biológicos , Miocardio/metabolismo , Ratas , Sirtuina 1/genética
10.
Biomolecules ; 10(3)2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32111073

RESUMEN

BACKGROUND: Matrix metalloproteinases (MMPs) are involved in vascular wall degradation, and drugs able to modulate MMP activity can be used to prevent or treat aneurysmal disease. In this study, we evaluated the effects of statins on MMP-2, MMP-9, and neutrophil gelatinase-associated lipocalin (NGAL) in both plasma and tissue in patients with aneurysmal disease. METHODS: We performed a prospective, single-blind, multicenter, control group clinical drug trial on 184 patients of both sexes >18 years old with a diagnosis of arterial aneurysmal disease. Enrolled patients were divided into two groups: Group I under statin treatment and Group II not taking statins. In addition, 122 patients without aneurysmal disease and under statin treatment were enrolled as a control group (Group III). The expression of MMPs and NGAL in plasma was evaluated using ELISA, while their expression in endothelial tissues was evaluated using Western blot. RESULTS: The ELISA test revealed greater plasma levels (p < 0.01) of MMPs and NGAL in Groups I and II vs. Group III. Western blot analysis showed higher expression (p < 0.01) of MMPs and NGAL in Group II vs. Group I, and this increase was significantly higher (p < 0.01) in patients treated with low potency statins compared to high potency ones. CONCLUSIONS: MMPs and NGAL seem to play a major role in the development of aneurysms, and their modulation by statins suggests that these drugs could be used to prevent arterial aneurysmal disease.


Asunto(s)
Aneurisma/tratamiento farmacológico , Aneurisma/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Lipocalina 2/análisis , Metaloproteinasa 2 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Anciano , Aneurisma/sangre , Femenino , Humanos , Lipocalina 2/sangre , Masculino , Metaloproteinasa 2 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/sangre , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos
11.
Ultrasound Med Biol ; 34(3): 370-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17935862

RESUMEN

The aim of our study was to determine if strain (S) and strain rate (SR) imaging are more sensitive indices with respect to standard echocardiographic parameters to assess cardiac function in an experimental model of doxorubicin (DOX)-induced cardiomyopathy. DOX was administered intraperitoneally 4x/wk at the dose of 1.25 mg/kg/d over four weeks in Wistar rats (n = 26). Other 14 Wistar rats were used as controls. Echocardiographic studies were performed before DOX treatment (baseline), after two and four weeks of treatment. At two and four weeks of DOX treatment, rat hearts were collected for histologic analysis. After two weeks of DOX treatment, there were no significant changes in standard echocardiographic parameters and in myocardial velocities, but after four weeks of treatment, ejection fraction significantly decreased and left ventricle dimensions significantly increased. Also, myocardial velocities were significantly reduced after four weeks of treatment. Conversely, S and SR values changed significantly already after two weeks of treatment. At baseline, S and SR values were 27 +/- 7% and 7.4 +/- 1.7, respectively, and they significantly decreased to 12 +/- 6% (p < 0.0001) and 6 +/- 1.5 (p < 0.005) at two weeks of treatment. These changes were also significant compared with control rat parameters (p < 0.001). At four weeks of DOX treatment, a progressive worsening occurred. Strain and SR further significantly decreased to 10 +/- 4% (p < 0.0001 vs. baseline) and 4.6 +/- 1.6 (p < 0.0001 vs. baseline), respectively. These changes were supported by histologic findings. Indeed, damage in myocardial tissue was demonstrated by histology already after two weeks of DOX treatment, with a damage progression after four weeks of treatment. Our data demonstrate that S and SR imaging are more sensitive indices in identifying early myocardial systolic changes induced by DOX than standard echocardiographic parameters and myocardial velocities. We believe that our model will be very supportive to further assess these new diagnostic strategies to provide a precocious diagnosis of LV dysfunction with a unique opportunity to initiate preventive cardioactive therapy. (E-mail: giodisal@yahoo.it).


Asunto(s)
Ecocardiografía Doppler en Color/métodos , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/diagnóstico por imagen , Animales , Antraciclinas , Progresión de la Enfermedad , Doxorrubicina , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Modelos Animales , Contracción Miocárdica , Miocardio/patología , Ratas , Ratas Wistar , Procesamiento de Señales Asistido por Computador , Volumen Sistólico , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
12.
Stem Cells Int ; 2018: 9492038, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29731780

RESUMEN

Pulmonary emphysema is a respiratory condition characterized by alveolar destruction that leads to airflow limitation and reduced lung function. Although with extensive research, the pathophysiology of emphysema is poorly understood and effective treatments are still missing. Evidence suggests that mesenchymal stem cells (MSCs) possess the ability to engraft the injured tissues and induce repair via a paracrine effect. Thus, the aim of this study was to test the effects of the intratracheal administration of lung-derived mouse MSCs in a model of elastase-induced emphysema. Pulmonary function (static lung compliance) showed an increased stiffness induced by elastase, while morphometric findings (mean linear intercept and tissue/alveolar area) confirmed the severity of alveolar disruption. Contrarily, MSC administration partially restored lung elasticity and alveolar architecture. In the absence of evidence that MSCs acquired epithelial phenotype, we detected an increased proliferative activity of aquaporin 5- and surfactant protein C-positive lung cells, suggesting MSC-driven paracrine mechanisms. The data indicate the mediation of hepatocyte growth factor in amplifying MSC-driven tissue response after injury. Our study shed light on supportive properties of lung-derived MSCs, although the full identification of mechanisms orchestrated by MSCs and responsible for epithelial repair after injury is a critical aspect yet to be achieved.

13.
Br J Pharmacol ; 174(22): 4070-4086, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27922176

RESUMEN

BACKGROUND AND PURPOSE: Heart failure with preserved ejection fraction (HFpEF) is a systemic syndrome driven by co-morbidities, and its pathophysiology is poorly understood. Several studies suggesting that dipeptidyl peptidase 4 (DPP4) might be involved in the pathophysiology of heart failure have prompted experimental and clinical investigations of DPP4 inhibitors in the cardiovascular system. Here we have investigated whether the DPP4 inhibitor sitagliptin affected the progression of HFpEF independently of its effects on glycaemia. EXPERIMENTAL APPROACH: Seven-week-old Dahl salt-sensitive rats were fed a high-salt diet for 5 weeks to induce hypertension. Then the rats continued with the high-salt diet and were treated with either sitagliptin (10 mg·kg-1 ) or vehicle for the following 8 weeks. Blood pressure and cardiac function were measured in vivo. Histochemical and molecular biology analyses of myocardium were used to assay cytokines, fibrotic markers, DPP4 and glucagon-like peptide-1 (GLP-1)/GLP-1 receptor. KEY RESULTS: Treatment with sitagliptin attenuated diastolic dysfunction, reduced mortality and reduced cardiac DPP4 activity, along with increased circulating GLP-1 and myocardial expression of GLP-1 receptors. Myocardial levels of pro-inflammatory cytokines (TNF-α, IL-6 and CCL2) were reduced. Sitagliptin treatment decreased the levels of endothelial NOS monomer, responsible for generation of ROS, while the amount of NO-producing dimeric form increased. Markers of oxidative and nitrosative stress were decreased. Moreover, increased collagen deposition and activation of pro-fibrotic signalling, inducing elevated myocardial stiffness, were attenuated by sitagliptin treatment. CONCLUSIONS AND IMPLICATIONS: Sitagliptin positively modulated active relaxation and passive diastolic compliance by decreasing inflammation-related endothelial dysfunction and fibrosis, associated with HFpEF. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Asunto(s)
Antiinflamatorios/uso terapéutico , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Fosfato de Sitagliptina/uso terapéutico , Animales , Antiinflamatorios/farmacología , Presión Sanguínea/efectos de los fármacos , Diástole/efectos de los fármacos , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Fibrosis , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Corazón/efectos de los fármacos , Corazón/fisiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Miocardio/patología , Óxido Nítrico/metabolismo , Ratas Endogámicas Dahl , Fosfato de Sitagliptina/farmacología , Volumen Sistólico
14.
Br J Pharmacol ; 174(21): 3696-3712, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28320043

RESUMEN

BACKGROUND AND PURPOSE: Doxorubicin is a highly effective anticancer drug, but its clinical application is hampered by cardiotoxicity. Asymptomatic diastolic dysfunction can be the earliest manifestation of doxorubicin cardiotoxicity. Therefore, a search for therapeutic intervention that can interfere with early manifestations and possibly prevent later development of cardiotoxicity is warranted. Increased doxorubicin-dependent ROS may explain, in part, Ca2+ and Na+ overload that contributes to diastolic dysfunction and development of heart failure. Therefore, we tested whether the administration of ranolazine, a selective blocker of late Na+ current, immediately after completing doxorubicin therapy, could affect diastolic dysfunction and interfere with the progression of functional decline. EXPERIMENTAL APPROACH: Fischer 344 rats received a cumulative dose of doxorubicin of 15 mg·kg-1 over a period of 2 weeks. After the assessment of diastolic dysfunction, the animals were treated with ranolazine (80 mg·kg-1 , daily) for the following 4 weeks. KEY RESULTS: While diastolic and systolic function progressively deteriorated in doxorubicin-treated animals, treatment with ranolazine relieved diastolic dysfunction and prevented worsening of systolic function, decreasing mortality. Ranolazine lowered myocardial NADPH oxidase 2 expression and oxidative/nitrative stress. Expression of the Na+ /Ca2+ exchanger 1 and Nav 1.5 channels was reduced and of the sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase 2 protein was increased. In addition, ranolazine lowered doxorubicin-induced hyper-phosphorylation and oxidation of Ca2+ /calmodulin-dependent protein kinase II, and decreased myocardial fibrosis. CONCLUSIONS AND IMPLICATIONS: Ranolazine, by the increased Na+ influx, induced by doxorubicin, altered cardiac Ca2+ and Na+ handling and attenuated diastolic dysfunction induced by doxorubicin, thus preventing the progression of cardiomyopathy. LINKED ARTICLES: This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.


Asunto(s)
Doxorrubicina/toxicidad , Ranolazina/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Disfunción Ventricular Izquierda/prevención & control , Animales , Antibióticos Antineoplásicos/toxicidad , Calcio/metabolismo , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Estrés Nitrosativo/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Sodio/metabolismo , Disfunción Ventricular Izquierda/inducido químicamente
15.
Diabetes ; 54(3): 803-10, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15734859

RESUMEN

This study investigated the role of heme oxygenase (HO)-1 in the cardiac tissue injury of acute ischemia/reperfusion (I/R) in diabetic streptozotocin (STZ)-induced hyperglycemic rats. The effects of 1) hemin, an inducer of HO expression and activity, and 2) zinc protoporphyrin IX (ZnPP-IX), an inhibitor of HO activity, have also been investigated on the tissue injury by I/R and some mediators released in these circumstances. STZ hyperglycemic rats had impaired levels of HO-1 within the cardiac tissue and increased myocardial infarct size (IS) following I/R, as compared with the nondiabetic rats. In these rats, administration of hemin 4 mg/kg 18 h before I/R increases the levels of HO-1 within the tissue. However, the values of HO-1 assayed in these circumstances were significantly lower (P < 0.01) than those assayed in nondiabetic animals subjected to the same procedures; IS was much more extended (P < 0.01) than in the parent nondiabetic group. STZ hyperglycemic rats also predisposed the heart to produce high levels of the cytokines interleukin (IL)-1beta and CXCL8. Subsequent I/R further increased (P < 0.01) the cytokine production, an effect partly prevented by hemin treatment. This recovered the huge number of infiltrated polymorphonuclear (PMN) leukocytes within the cardiac tissue associated with the STZ hyperglycemic state and I/R damage.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Proteínas de Choque Térmico/fisiología , Hiperglucemia/fisiopatología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Miocardio/enzimología , Oxigenasas/fisiología , Animales , Antígeno CD11b/fisiología , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/enzimología , Expresión Génica , Proteínas de Choque Térmico/metabolismo , Hemo Oxigenasa (Desciclizante) , Hemina/fisiología , Hiperglucemia/enzimología , Hiperglucemia/patología , Leucocitos Mononucleares , Masculino , Infarto del Miocardio/sangre , Infarto del Miocardio/enzimología , Daño por Reperfusión Miocárdica/sangre , Daño por Reperfusión Miocárdica/enzimología , Miocardio/patología , Oxigenasas/metabolismo , Protoporfirinas/fisiología , Ratas , Ratas Sprague-Dawley
16.
Cardiooncology ; 2(1): 2, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-33530140

RESUMEN

The cardiotoxicity of doxorubicin is becoming an interdisciplinary point of interest given a growing population of cancer survivors. The complex and not completely understood pathogenesis of this complication makes difficult to design successful preventive or curative measures. Although cardiomyocyte has been considered a classical cellular target, other cells including various types of undifferentiated cells are involved in myocardial homeostasis. Such perspective may shed light on previously unrecognized aspects of cardiotoxicity and promote new experimental and clinical cardioprotective strategies. In this review, different cellular targets of doxorubicin are discussed with the focus on cardiac progenitor cells, oxidative stress, DNA damage, senescence and apoptosis all of which contribute to their compromised functional properties.

17.
PLoS One ; 11(7): e0158746, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27434719

RESUMEN

BACKGROUND: The need for new options for chronic lung diseases promotes the research on stem cells for lung repair. Bone marrow-derived mesenchymal stem cells (MSCs) can modulate lung inflammation, but the data on cellular processes involved in early airway remodeling and the potential involvement of neuropeptides are scarce. OBJECTIVES: To elucidate the mechanisms by which local administration of MSCs interferes with pathophysiological features of airway hyperresponsiveness in an animal model. METHODS: GFP-tagged mouse MSCs were intratracheally delivered in the ovalbumin mouse model with subsequent functional tests, the analysis of cytokine levels, neuropeptide expression and histological evaluation of MSCs fate and airway pathology. Additionally, MSCs were exposed to pro-inflammatory factors in vitro. RESULTS: Functional improvement was observed after MSC administration. Although MSCs did not adopt lung cell phenotypes, cell therapy positively affected airway remodeling reducing the hyperplastic phase of the gain in bronchial smooth muscle mass, decreasing the proliferation of epithelium in which mucus metaplasia was also lowered. Decrease of interleukin-4, interleukin-5, interleukin-13 and increase of interleukin-10 in bronchoalveolar lavage was also observed. Exposed to pro-inflammatory cytokines, MSCs upregulated indoleamine 2,3-dioxygenase. Moreover, asthma-related in vivo upregulation of pro-inflammatory neurokinin 1 and neurokinin 2 receptors was counteracted by MSCs that also determined a partial restoration of VIP, a neuropeptide with anti-inflammatory properties. CONCLUSION: Intratracheally administered MSCs positively modulate airway remodeling, reduce inflammation and improve function, demonstrating their ability to promote tissue homeostasis in the course of experimental allergic asthma. Because of a limited tissue retention, the functional impact of MSCs may be attributed to their immunomodulatory response combined with the interference of neuropeptide system activation and tissue remodeling.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Receptores de Neuroquinina-1/inmunología , Receptores de Neuroquinina-2/inmunología , Hipersensibilidad Respiratoria/terapia , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Intubación Intratraqueal , Pulmón/inmunología , Pulmón/patología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-2/genética , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología
18.
Int J Cardiol ; 205: 99-110, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26730840

RESUMEN

BACKGROUND: Doxorubicin (DOXO) is an effective anti-neoplastic drug but its clinical benefits are hampered by cardiotoxicity. Oxidative stress, apoptosis and myocardial fibrosis mediate the anthracycline cardiomyopathy. ROS trigger TGF-ß pathway that activates cardiac fibroblasts promoting fibrosis. Myocardial stiffness contributes to diastolic dysfunction, less studied aspect of anthracycline cardiomyopathy. Considering the role of SIRT1 in the inhibition of the TGF-ß/SMAD3 pathway, resveratrol (RES), a SIRT1 activator, might improve cardiac function by interfering with the development of cardiac fibrosis in a model of DOXO-induced cardiomyopathy. METHODS: F344 rats received a cumulative dose of 15 mg/kg of DOXO in 2 weeks or DOXO+RES (DOXO and RES, 2.5mg/kg/day, concomitantly for 2 weeks and then RES alone for 1 more week). The effects of RES on cardiac fibroblasts were also tested in vitro. RESULTS: Along with systolic dysfunction, DOXO was also responsible of diastolic abnormalities. Myocardial stiffness correlated with fibroblast activation and collagen deposition. DOXO+RES co-treatment significantly improved ± dP/dt and, more interestingly, ameliorated end-diastolic pressure/volume relationship. Treatment with RES resulted in reduced fibrosis and fibroblast activation and, most importantly, the mortality rate was significantly reduced in DOXO+RES group. Fibroblasts isolated from DOXO+RES-treated rats, in which SIRT1 was upregulated, showed decreased levels of TGF-ß and pSMAD3/SMAD3 when compared to cells isolated from DOXO-exposed hearts. CONCLUSIONS: Our findings reveal a key role of SIRT1 in supporting animal survival and functional parameters of the heart. SIRT1 activation by interfering with fibrogenesis can improve relaxation properties of myocardium and attenuate myocardial remodeling related to chemotherapy.


Asunto(s)
Cardiomiopatías/metabolismo , Cardiomiopatías/prevención & control , Diástole/efectos de los fármacos , Doxorrubicina/toxicidad , Sirtuina 1/metabolismo , Estilbenos/uso terapéutico , Animales , Antraciclinas/toxicidad , Antibióticos Antineoplásicos/toxicidad , Cardiomiopatías/inducido químicamente , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibrosis , Ratas , Ratas Endogámicas F344 , Resveratrol , Estilbenos/farmacología
19.
Oncotarget ; 7(38): 62312-62326, 2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27694688

RESUMEN

New strategies to prevent and early detect the cardiotoxic effects of the anticancer drug doxorubicin (DOXO) are required. MicroRNAs emerged as potential diagnostic, therapeutic and prognostic approaches in cardiovascular diseases. MiR-34a has a role in cardiac dysfunction and ageing and is involved in several cellular processes associated with DOXO cardiotoxicity. Our in vitro and in vivo results indicated that after DOXO exposure the levels of miR-34a are enhanced in cardiac cells, including Cardiac Progenitor Cells (CPCs). Since one of the determining event responsible for the initiation and evolution of the DOXO toxicity arises at the level of the CPC compartment, we evaluated if miR-34a pharmacological inhibition in these cells ameliorates the detrimental aftermath of the drug. AntimiR-34a has beneficial consequences on vitality, proliferation, apoptosis and senescence of DOXO-treated rat CPC. These effects are mediated by an increase of prosurvival miR-34a targets Bcl-2 and SIRT1, accompanied by a decrease of acetylated-p53 and p16INK4a. Importantly, miR-34a silencing also reduces the release of this miRNA from DOXO-exposed rCPCs, decreasing its negative paracrine effects on other rat cardiac cells. In conclusion, the silencing of miR-34a could represent a future therapeutic option for cardioprotection in DOXO toxicity and at the same time, it could be considered as a circulating biomarker for anthracycline-induced cardiac damage.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiomiopatías/sangre , Doxorrubicina/efectos adversos , Corazón/efectos de los fármacos , MicroARNs/metabolismo , Mioblastos Cardíacos/metabolismo , Acetilación , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/análisis , Biomarcadores/metabolismo , Cardiomiopatías/inducido químicamente , Cardiomiopatías/genética , Cardiotoxicidad/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales , Femenino , MicroARNs/antagonistas & inhibidores , MicroARNs/sangre , Mioblastos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Endogámicas F344 , Reacción en Cadena en Tiempo Real de la Polimerasa , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
20.
Int J Cardiol ; 217: 69-79, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-27179211

RESUMEN

BACKGROUND: To investigate the effects of chronic administration of ranolazine (RAN) on experimental model of heart failure with preserved ejection fraction. METHODS: Seven-weeks old Dahl salt-sensitive rats were fed a high salt diet for 5weeks to induce hypertension. Afterwards, rats continued with a high salt diet and were administered either with vehicle or RAN (20mg/kg/die, ip) for the following 8weeks. Control rats were maintained on a low salt diet. RESULTS: While systolic parameters were not altered, diastolic parameters were changed in high salt animals. Hemodynamic analysis showed a decreased dP/dt min, increased LVEDP, longer time constant and steeper slope of the end-diastolic pressure-volume relationship. Treatment with RAN attenuated these alterations and determined a reduction in mortality. Additionally, the magnitude of myocardial hypertrophy and activation of PI3K/Akt pathway were reduced. Alteration in diastolic compliance as a consequence of elevated myocardial stiffness was confirmed by an increase of collagen deposition and activation of pro-fibrotic TGF-ß/SMAD3/CTGF signaling. These effects were counteracted by RAN. High salt rats had a decrease in SERCA2 and an increase in Na(+)/Ca(2+) exchanger (NCX). Treatment with RAN reduced NCX expression and determined an increment of SERCA2. Moreover, the levels of nitrotyrosine and oxidized dyhydroethidium were higher in high salt rats. RAN induced a decrement of oxidative stress, supporting the concept that reduction in ROS may mediate beneficial effects. CONCLUSIONS: Our findings support the possibility that diastolic dysfunction can be attenuated by RAN, indicating its ability to affect active relaxation and passive diastolic compliance.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Insuficiencia Cardíaca/tratamiento farmacológico , Ranolazina/administración & dosificación , Volumen Sistólico/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Esquema de Medicación , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas Dahl , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA