Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
2.
Biol. Res ; 54: 3-3, 2021. graf, ilus
Article in English | LILACS | ID: biblio-1505792

ABSTRACT

BACKGROUND: Testosterone regulates nutrient and energy balance to maintain protein synthesis and metabolism in cardiomyocytes, but supraphysiological concentrations induce cardiac hypertrophy. Previously, we determined that testosterone increased glucose uptake­via AMP-activated protein kinase (AMPK)­after acute treatment in cardiomyocytes. However, whether elevated glucose uptake is involved in long-term changes of glucose metabolism or is required during cardiomyocyte growth remained unknown. In this study, we hypothesized that glucose uptake and glycolysis increase in testosterone-treated cardiomyocytes through AMPK and androgen receptor (AR). METHODS: Cultured cardiomyocytes were stimulated with 100 nM testosterone for 24 h, and hypertrophy was verified by increased cell size and mRNA levels of ß-myosin heavy chain (ß-mhc). Glucose uptake was assessed by 2-NBDG. Glycolysis and glycolytic capacity were determined by measuring extracellular acidification rate (ECAR). RESULTS: Testosterone induced cardiomyocyte hypertrophy that was accompanied by increased glucose uptake, glycolysis enhancement and upregulated mRNA expression of hexokinase 2. In addition, testosterone increased AMPK phosphorylation (Thr172), while inhibition of both AMPK and AR blocked glycolysis and cardiomyocyte hypertrophy induced by testosterone. Moreover, testosterone supplementation in adult male rats by 5 weeks induced cardiac hypertrophy and upregulated ß-mhc, Hk2 and Pfk2 mRNA levels. CONCLUSION: These results indicate that testosterone stimulates glucose metabolism by activation of AMPK and AR signaling which are critical to induce cardiomyocyte hypertrophy.


Subject(s)
Animals , Male , Rats , Testosterone/pharmacology , Receptors, Androgen/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Signal Transduction , Cells, Cultured , Hypertrophy , Myocardium/pathology
3.
Rev. chil. cardiol ; 39(1): 66-74, abr. 2020. graf
Article in Spanish | LILACS | ID: biblio-1115452

ABSTRACT

En 31 de diciembre del 2019 la Organización Mundial de la Salud fue informada por las autoridades sanitarias chinas de la aparición de casos de neumonía de origen desconocido en la ciudad de Wuhan en China. El 7 de Enero de 2020, científicos chinos identificaron a un nuevo coronavirus (temporalmente designado como "2019-nCoV") como el agente etiológico de la enfermedad denominada COVID-19. La secuenciación del genoma del nuevo coronavirus mostró gran similitud con el coronavirus (Covid-1 o SARS-CoV) causante del síndrome respiratorio agudo severo (SARS), ocurrido también en China entre los años 2002-2003. Por este motivo, 2019-nCoV se rebautizó como SARS-CoV-2 (Severe Acute Respiratory Syndrome Corona Virus-2) y a la fecha es responsable de la actual y grave pandemia que está ocasionando impactos sanitarios y socio-económicos a escala global. Las investigaciones con SARS-CoV establecieron que este virus ingresa a nuestras células utilizando como receptor a la enzima convertidora de angiotensina tipo 2 (ECA 2 o en inglés ACE-2: "angiotensin converting enzyme type 2"). Dado este antecedente también se confirmó que SARS-CoV-2 también utiliza esta misma enzima ya que no se habla de un mecanismo en si para ingresar a sus células blanco, especialmente a nivel de nuestro sistema respiratorio. ECA-2 es una proteasa integrante del sistema renina angiotensina "alterno o no canónico" con importantes acciones regulatorias sobre los sistemas cardiovascular, renal y pulmonar, entre otros. En este contexto, ha surgido preocupación tanto por clínicos como los propios pacientes respecto al estado de pacientes hipertensos con COVID-19 y su vulnerabilidad a infectarse con SARS-CoV-2 dado que algunos trabajos han planteado que ciertos polimorfismos en el gen ECA-2 asociados a hipertensión arterial podrían determinar una mayor expresión de ECA-2. Además, estudios preclínicos han sugerido que ciertos fármacos antihipertensivos (principalmente, inhibidores de ECA y antagonistas del receptor para angiotensina II subtipo 1) también podrían estimular una mayor expresión de ECA-2. Esta revisión tiene por objetivo presentar y discutir los antecedentes en el estado del arte respecto a esta reciente problemática. El análisis crítico de los presentes antecedentes permite concluir que no existe evidencia clínica sólida que permita afirmar que el uso de medicamentos antihipertensivos genere una mayor vulnerabilidad a la infección con SARS-CoV-2. Por lo tanto no se debe descontinuar su uso en pacientes hipertensos en riesgo de infección a SARS-CoV-2 o que padezcan COVID-19.


In December 2019, a new type of coronavirus emerged in the city of Wuhan, China. This novel virus has unleashed a pandemic that has inflicted a considerable impact on public health and the economy and has therefore become a severe concern worldwide. This new virus -named SARS-CoV-2has been rapidly investigated in order to create knowledge aimed at achieving its control. Comparative studies with SARS-CoV virus, responsible for the 2002-2003 pandemic, suggest that SARS-CoV-2 requires the same receptor to bind and infect cells: angiotensin converting enzyme 2 (ACE-2). This hypothesis has been thoroughly supported by a variety of in vitro research and is currently considered a potential therapeutic target. ACE-2 is part of the counter-regulatory renin-angiotensin system, exerting effects in pulmonary, renal and cardiovascular systems. In this context, concerns have arisen in regards to the vulnerability of hypertensive patients against COVID-19, given that there is evidence that may suggest that polymorphisms associated to hypertension may increase the expression of ACE-2. Moreover, preclinical studies have shown that antihypertensive drugs may increase the expression of this enzyme. In this review article, we present the current state of the art on this polemic topic. Our critical analysis suggest that there is no robust clinical evidence supporting the hypothesis that the use of antihypertensive drugs can increase vulnerability to infection with SARS-CoV-2. Therefore, we recommend that the use of these therapeutic agents should not be discontinued in hypertensive patients in risk to or suffering COVID-19.


Subject(s)
Humans , Pneumonia, Viral/complications , Coronavirus Infections/complications , Hypertension/complications , Hypertension/drug therapy , Antihypertensive Agents/therapeutic use , Pneumonia, Viral/metabolism , Renin-Angiotensin System , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Coronavirus Infections/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Pandemics , Betacoronavirus/metabolism , Hypertension/metabolism
4.
Rev. chil. cardiol ; 38(3): 225-235, dic. 2019. graf
Article in Spanish | LILACS | ID: biblio-1058068

ABSTRACT

RESUMEN: El Premio Nobel 2019 en Fisiología-Medicina se confirió a los Profesores Gregg Semenza, William Kaelin y Sir Peter Ratcliffe por sus investigaciones en la maquinaria molecular que regula la expresión de genes sensibles a los cambios en los niveles de oxígeno. La síntesis de eritropoyetina inducida por la disminución de los niveles sanguíneos de oxígeno condujo al estudio del gen de la eritropoyetina y descubrimiento de los elementos de respuesta a hipoxia (HRE) en la región promotora y posteriormente al factor transcripcional inducible por hipoxia tipo 1 (HIF-1). Este factor consta de dos subunidades: HIF-1α, sensible al oxígeno, y HIF-1β, expresada constitutivamente. HIF1 activa la transcripción de genes que codifican enzimas, transportadores y proteínas mitocondriales que disminuyen la utilización de oxígeno al cambiar el metabolismo oxidativo al metabolismo glicolítico y además aquellos involucrados en la angiogénesis y diferenciación celular. Las investigaciones paralelas en la enfermedad von Hippel-Lindau (VHL), un desorden autosómico dominante, permitieron descubrir el mecanismo de degradación de HIF1 en condiciones de normoxia y como se estabiliza bajo hipoxia. El impacto de HIF en clínica radica en el establecimiento de nuevas dianas terapéuticas para combatir la anemia y diversas enfermedades cardiovasculares. HIF promueve la angiogénesis a través de la expresión del factor de crecimiento vascular endotelial (VEGF), agente cardioprotector con potencial para tratar la isquemia/reperfusión, hipertrofia patológica e insuficiencia cardíaca.


ABSTRACT: The Nobel Prize in Physiology-Medicine was awarded to Drs. Gregg Semenza, William Kaelin and Sir Peter Ratcliffe for their research in the molecular machinery that regulates the expression of genes sensitive to the change in oxygen levels. The synthesis of erythropoietin induced by the decrease levels of oxygen in the blood led to investigate the promoter of the erythropoietin gene where the so-called hypoxia response elements (HRE) were described. Semenza et al. described a protein that binds to HREs and called it hypoxia-inducible transcriptional factor (HIF) that regulates gene expression among those involved in angiogenesis, cell differentiation and glycolytic enzymes. HIF presents two oxygen-sensitive subunits HIF-1α and HIF-1β constitutively expressed. In parallel, Kaelin et al. investigated von Hippel-Lindau disease (VHL), an autosomal dominant disorder, discovering a mutation of this protein generated a behavior similar to hypoxia. The impact of HIF-1α lies in the search for new strategies such as hydrolase inhibitors to combat prevalent diseases, including anemia and cardiovascular diseases These compounds promote the expression of vascular endothelial growth factor (VEGF), a cardioprotective agent with potential use in pre- and post-conditioning therapy, cardiac hypertrophy and heart failure.


Subject(s)
Humans , Male , Female , Cardiovascular Diseases , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit , Receptors, Vascular Endothelial Growth Factor , Angiogenesis Inducing Agents , Heart Failure , von Hippel-Lindau Disease/genetics , Hypoxia , Nobel Prize
5.
Rev. chil. cardiol ; 38(1): 37-45, abr. 2019. tab, graf
Article in Spanish | LILACS | ID: biblio-1003636

ABSTRACT

Resumen: Introducción: Atletas altamente entrenados muestran cambios cardíacos estructurales como adaptación a la sobrecarga, producto del ejercicio repetitivo y extenuante. Se han evidenciado elevación de biomarcadores de remodelado y fibrosis miocárdica posterior al ejercicio intenso en atletas. Sin embargo, el comportamiento de estos biomarcadores según el nivel de entrenamiento previo no se ha evaluado. Objetivo: Investigar biomarcadores de fibrosis y función ventricular derecha en maratonistas con distinto nivel de entrenamiento previo. Métodos: Se incluyeron 36 maratonistas hombres, sanos, que completaron 42 km en la maratón de Santiago. Se dividieron según entrenamiento previo en dos grupos, Grupo 1 (G1): ≥100 km/semana y Grupo 2 (G2): <100 km/semana. Se realizó ecocardiografía transtorácica y se evaluaron niveles plasmáticos de galectina-3 y del propéptido amino terminal del procolágeno tipo III (PIIINP) en la semana previa a la carrera e inmediatamente posterior a ésta. Resultados: Posterior a la maratón, la función sistólica del ventrículo derecho disminuyó en el grupo G2 junto con un aumento significativo de los niveles plasmáticos de PIIIPNP (61±16 a 94±24 ng/mL, p=0,01). Estos cambios no se observaron en el grupo G1 (65 ± 11 a 90±29 ng/mL, p=0,10). Los niveles plasmáticos de galectina-3 aumentaron significativamente en ambos grupos posterior al ejercicio (6,8±2,2 a 19,7±4,9 ng/mL, p 0,012 y 6,0±1,1 a 19,4 ± 5,9 ng/mL, p 0,01) en los grupos G1 y G2, respectivamente). Conclusiones: Atletas con menor grado de entrenamiento, presentan posterior a una maratón un significativo aumento de productos de degradación del colágeno (PIIIPNP) asociado a disminución de la función del ventrículo derecho. Los niveles de galectina-3 plasmática aumentan significativamente en ambos grupos post-esfuerzo independiente del entrenamiento previo.


Abstracts: Introduction: Highly trained athletes show structural cardiac changes as adaptation to overload. Rise in remodeling biomarkers and myocardial fibrosis after intense exercise in athletes has been evidenced; however, the behavior of these biomarkers according to pre-competition training level has not been evaluated. Objective: To evaluate fibrosis biomarkers levels and right ventricle function in marathon runners according to their previous training level, in the period prior to a marathon race and immediately after it. Methods: Thirty-six healthy male marathon runners were included. Subjects were grouped according to their previous training level: Group 1 (G1): ≥100 km/week and Group 2 (G2): <100 km/week. Transthoracic echocardiography along with plasmatic levels of galectin-3 and amino terminal propeptide of type III procollagen (PIIINP) were measured one week previous and immediately after the marathon. Results: Post-effort right ventricle systolic function decreased in G2, together with a significant elevation of PIIIPNP (61±16 to 94±24 ng/mL, p=0.01). These changes were not observed in G1 (from 65±11 to 90±29 ng/mL, p=0.10). Plasma galectin-3 increased significantly in both groups immediately post-exercise (6.8±2.2 to 19.7±4.9 ng/mL, p=0.012, and 6.0±1.1 to 19.4±5.9 ng/mL, p=0.01, in G1 and G2. respectively). Conclusion: Less trained athletes evidenced higher post marathon levels of PIIIPNP which is associated with a decreased global right ventricle function. Plasma galectin-3 levels increased significantly after intense exertion regardless of the intensity of previous training.


Subject(s)
Humans , Male , Adult , Middle Aged , Running/physiology , Fibrosis/blood , Biomarkers/blood , Ventricular Function, Right , Heart Injuries/blood , Peptide Fragments/blood , Fibrosis/physiopathology , Exercise/physiology , Single-Blind Method , Chile , Prospective Studies , Longitudinal Studies , Ventricular Function, Left , Procollagen/blood , Galectin 3/blood , Athletes
6.
Rev. chil. cardiol ; 38(1): 54-63, abr. 2019. graf
Article in Spanish | LILACS | ID: biblio-1003638

ABSTRACT

Resumen: Las enfermedades cardiovasculares y el cáncer son enfermedades crónicas transmisibles culturalmente, y las dos causas principales de mortalidad en el mundo. Además del gran impacto sobre la mortalidad y morbilidad, estas enfermedades han mostrado un alto grado de relación entre ellas debido, entre otras razones, a que comparten factores de riesgo y mecanismos biológicos. La alta incidencia de enfermedad cardiovascular en pacientes con cáncer es un fenómeno conocido que ha orientado el desarrollo del campo interdisciplinario de la cardio-oncología. Sin embargo, en la última década han surgido evidencias que muestran el papel que desempeñan las enfermedades cardiovasculares en el desarrollo de cáncer. Un estudio reciente publicado por Meijers y cols, en agosto de 2018 en Circulation, mostró que la insuficiencia cardiaca post-infarto del miocardio contribuye significativamente al desarrollo del cáncer de colón, apoyando lo obtenido en estudios epidemiológicos anteriores. Este estudio también sugiere que el crecimiento tumoral podría producirse por factores secretados por el corazón insuficiente abriendo un amplio grupo de posibilidades de investigación en lo que sería un nuevo campo de la medicina cuyo propósito sería el desarrollo de nuevas estrategias para el seguimiento y tratamiento del cáncer en pacientes con enfermedades cardiovasculares. El presente artículo revisa los factores de riesgo, y mecanismos celulares y moleculares, que son comunes en las enfermedades cardiovasculares y el cáncer, la contribución del trabajo de Meijers y cols hacia un mayor entendimiento de la interrelación entre estas patologías y las perspectivas futuras con respecto a los nuevos hallazgos.


Abstracts: Cardiovascular diseases and cancer are culturally transmitted chronic diseases and the two main causes of death globally. In addition to their high morbidity and mortality, these diseases are closely related, due to their common risk factors and biological mechanisms. The high incidence of cardiovascular diseases in cancer patients is widely known phenomenon, which has oriented the development of the interdisciplinary field of cardio-oncology Nonetheless, there is emerging evidence in the last decade suggesting a potential role for cardiovascular diseases in the onset of cancer. A recent publication by Meijers et al in the scientific cardiovascular journal Circulation showed that heart failure significantly contributes to tumor growth, confirming previous epidemiological findings suggesting this hypothesis. Moreover, this study indicates that tumor growth may be stimulated by the secretion of factors from the failing heart, opening a wide spectrum of research areas in what may be suggested as a new field in medicine that would seek to develop new strategies to treat and prevent cancer in patients with cardiovascular diseases. This article will review shared risk factor and common cellular and molecular pathways in cardiovascular diseases and cancer, the contribution of Meijers et al to a better understanding of the connection of these diseases and future perspectives in light of the new evidence.


Subject(s)
Humans , Cardiovascular Diseases/epidemiology , Neoplasms/epidemiology , Risk Factors , Heart Failure/epidemiology
7.
Rev. chil. cardiol ; 37(2): 93-103, ago. 2018. tab, graf, ilus
Article in Spanish | LILACS | ID: biblio-959346

ABSTRACT

Resumen: Introducción: El ejercicio físico reduce la mortalidad cardiovascular y genera remodelado cardíaco. Altas cargas de entrenamiento pueden generar remodelado cardíaco adverso. Biomarcadores (BMC) de inflamación Interleukina 6 (IL-6) y de estrés oxidativo Malondialdehído (MDA), potencialmente pueden caracterizar la respuesta al esfuerzo. Objetivo: Evaluar actividad de IL-6 y MDA en respuesta a una maratón en atletas con distinto nivel de entrenamiento y remodelado cardíaco asociado. Sujetos y Métodos: Estudio prospectivo, simple ciego, incluyó 16 atletas que completaron la maratón de Santiago (42 k), separados según entrenamiento previo, grupo 1 (G1, n: 8): Alto ≥ 100 km/semana y grupo 2 (G2, n: 8): Bajo <100 km/semana). Se obtuvo pre y post maratón: niveles de IL-6, MDA y ecocardiografía Doppler transtorácica (ETT); cuantificando cámaras cardíacas izquierdas, derechas y deformación del ventrículo izquierdo (strain longitudinal). Se utilizaron las pruebas de Mann-Whitney, Wilcoxon y Kruskal-Wallis. Resultados: Edad G1: 38.13±7.18 años vs G2: 40.38±6.63 años (NS). Tiempo maratón G1: 185.75±14.87 min vs G2: 219.75±24.92 min (p<0.01). Masa del ventrículo izquierdo G1: 91±21 g/m2 vs G2: 73±12 g/m2 (p<0.01). Volumen aurícula izquierda G1: 39.4±12.6 ml/m2 vs 30.6±4.6 ml/m2 (p<0.01). FEVI G1: 55.8±3.3% vs G2: 58.6±6.7% (NS). MDA G1: PRE 0.17±0.13 uM/L, POST 0.67±0.59 uM/L, G2: PRE 0.29±0.24 uM/L, POST 1.01±1.15 uM/L (p<0.01). IL-6 G1: PRE 2.50±1.35 pg/ml, POST 93.91±27.23 pg/ml vs G2: PRE 4.65±5.89 pg/ml, POST 97.83±30.72 pg/ml (NS). Conclusión: El ejercicio físico aumenta los BMC de inflamación y estés oxidativo (IL-6, MDA). Un entrenamiento físico de alta intensidad disminuye la respuesta de estrés oxidativo y se asocia a un mayor remodelado cardíaco.


Abstract: Background: Exercise reduces cardiovascular mortality and generates cardiac remodeling. High training loads can induce adverse cardiac remodeling, and its associated cardiac remodeling. Therefore, interleukin 6 (IL 6) and malondialdehyde (MDA), biomarkers of inflammatory response and oxidative stress respectively, may have a role in stratifying this risk. Objective: To assess the activity of IL-6 and MDA in response to a marathon race in athletes with different previous training status. Subjects And Methods: Prospective, single-blind study involving 16 male athletes that finished the Santiago Marathon (42 k), allocated into two groups according to their previous training: Group 1 (G1, n: 8) with high training (≥ 100 km/weekly) and Group 2 (G2, n: 8) with low training (< 100 km/weekly). Before and after the race serum levels of IL-6, MDA and transthoracic Doppler echocardiography for cardiac chamber quantification and left ventricle deformation (longitudinal strain) were measured. Mann-Whitney, Wilcoxon, and Kruskal-Wallis tests were used to assess statistical significance. Results: Age G1: 38.13±7.18 years-old vs G2: 40.38±6.63 years-old (NS). Marathon finishing time G1: 185.75±14.87 min vs G2: 219.75±24.92 min (p<0.01). Left ventricle mass G1: 91±21 g/m2 vs G2: 73±12 g/m2 (p<0.01). Left atrium volume G1: 39.4±12.6 ml/m2 vs 30.6±4.6 ml/m2 (p<0.01). LVEF G1: 55.8±3.3% vs G2: 58.6±6.7% (NS). MDA G1: PRE 0.17±0.13 uM/L, POST 0.67±0.59 uM/L, G2: PRE 0.29±0.24 uM/L, POST 1.01±1.15 uM/L (p<0.01). IL-6 G1: PRE 2.50±1.35 pg/ml, POST 93.91±27.23 vs G2: PRE 4.65±5.89 pg/ml, POST 97.83±30.72 pg/ml (NS). Conclusion: Physical exercise generates a rise in biomarkers (IL-6, MDA). Athletes with high-intensity training level have a diminished oxidative stress response post effort and greater cardiac remodeling.


Subject(s)
Humans , Male , Adolescent , Adult , Middle Aged , Young Adult , Interleukin-6/metabolism , Ventricular Remodeling/physiology , Exercise Therapy/methods , Malondialdehyde/metabolism , Echocardiography , Biomarkers , Single-Blind Method , Prospective Studies , Oxidative Stress/physiology , Athletes , Heart Ventricles/diagnostic imaging
8.
Rev. chil. cardiol ; 37(1): 42-54, abr. 2018. tab, ilus
Article in Spanish | LILACS | ID: biblio-959338

ABSTRACT

Resumen: En los últimos años, la diabetes mellitus tipo 2 (DM2) ha evolucionado en forma epidémica, experimentando un rápido crecimiento y afectando a millones de individuos a nivel mundial. La cardiopatía isquémica es la principal causa de mortalidad en los pacientes diabéticos, quienes poseen un mayor riesgo cardiovascular respecto a los no diabéticos. La DM2 y la cardiopatía isquémica se caracterizan por ser prevenibles, sin embargo, existen diversos factores de riesgo comunes que contribuyen a su desarrollo. Los mecanismos que explican la ateroesclerosis acelerada y el incremento de riesgo de enfermedades cardiovasculares en los pacientes diabéticos tipo 2 incluyen a la hiperglicemia, dislipidemia y la inflamación del endotelio vascular. La diabetes es resultado de una interacción compleja entre la genética y el medio ambiente. Recientemente se han descrito varios genes implicados en el desarrollo de la diabetes y cardiopatía isquémica y que podrían significar nuevas opciones terapéuticas. En este artículo se revisa la relación entre ambas patologías, los mecanismos moleculares y el descubrimiento de factores de riesgo genéticos comunes y su implicancia en el desarrollo de nuevos blancos terapéuticos.


Abstracts: In recent years, type 2 diabetes mellitus has evolved as a rapidly increasing epidemic and affects millions of people worldwide. Ischemic heart disease (IHD) is the main cause of death among diabetic patients, who have a higher cardiovascular risk than non-diabetics. Both, DM2 and IHD are characterized by being preventable, however there are several common risk factors that contribute to their development. The mechanisms that explain accelerated atherosclerosis and increased risk of cardiovascular diseases in patients with type 2 diabetes mellitus include damage by hyperglycemia, dyslipidemia and inflammation on vascular endothelium. Diabetes is the result of a complex interaction between genetics and the environment, recently, several genes have been identified that appear to be involved in diabetes and ischemic heart disease that could explain its relationship and serve as new therapeutic possibilities. In this article, we review the relationship between diabetes and ischemic heart disease, the molecular mechanisms and the discovery of genetic risk factors common to both diseases and their implication in the development of new therapeutic targets.


Subject(s)
Humans , Myocardial Ischemia/etiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/genetics , Polymorphism, Genetic/genetics , Genetic Therapy , Myocardial Ischemia/physiopathology , Myocardial Ischemia/genetics , Myocardial Ischemia/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Dyslipidemias/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hyperglycemia/physiopathology , Metformin/therapeutic use
9.
Rev. méd. Chile ; 146(1): 68-77, ene. 2018. tab, graf
Article in Spanish | LILACS | ID: biblio-902623

ABSTRACT

Recently, we have witnessed major improvements in cancer treatment. Early diagnosis and development of new therapies have reduced cancer-related mortality. However, these new therapies, along with greater patient survival, are associated with an increase in untoward effects, particularly in the cardiovascular system. Although cardiotoxicity induced by oncologic treatments affects predominantly the myocardium, it can also involve other structures of the cardiovascular system, becoming one of the main causes of morbidity and mortality in those who survive cancer. The main objective of cardio-oncology is to achieve the maximum benefits of oncologic treatments while minimizing their deleterious cardiovascular effects. It harbors the stratification of patients at risk of cardiotoxicity, the implementation of diagnostic tools (imaging techniques and biomarkers) for early diagnosis, preventive strategies and early treatment options for the complications. Herein, we discuss the basic knowledge for the implementation of cardio-oncology units and their role in the management of cancer patients, the diagnostic tools available to detect cardiotoxicity and the present therapeutic options.


Subject(s)
Humans , Radiotherapy/adverse effects , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Antineoplastic Agents/adverse effects , Biomarkers , Risk Factors , Program Development , Neoplasms/complications , Neoplasms/drug therapy , Antineoplastic Agents/classification
10.
Rev. chil. cardiol ; 36(2): 89-96, 2017. ilus, tab
Article in Spanish | LILACS | ID: biblio-899572

ABSTRACT

Introducción: La miopatía y fibrosis auricular representan el sustrato protrombótico y proarrítmico en pacientes con fibrilación auricular (FA). Estudios recientes muestran relación entre el strain auricular izquierdo (SAI), eventos cardiovasculares y recurrencia en pacientes con FA. La asociación entre SAI y bio-marcadores cardíacos como predictores de accidente cerebrovascular silente (ACVs) en pacientes con FA de reciente comienzo (FArc) no ha sido estudiada. Objetivo: Determinar si la asociación entre SAI y biomarcadores cardíacos contribuye a la predicción de ACV en pacientes con FArc. Métodos: Se realizó un estudio prospectivo que permitió reclutar 57 pacientes con FArc (primer episodio de < de 8 semanas de evolución). Obtenido consentimiento informado (CI) se realizó recolección de datos clínicos y muestras de sangre para determinación de Pro-BNP, Dimero-D y GDF-15. Se realizó resonancia nuclear magnética cerebral (RNMc) y ecocardiograma transtorácico (ETT) durante los primeros 3 días de inclusión y en ritmo sinusal. Para la evaluación de SAI se consideró la curva de deflexión positiva durante la sístole ventricular (SAIs), derivada de speckle tracking, considerando el promedio de 5 ciclos. Se utilizó Mann Whitney U test y Spearman Rho para análisis estadístico. Resultados: La edad promedio fue 70±8,2 años y el 70% fueron hombres. El CHA2DS2-VASc score promedio fue 3,1±1 y el promedio de pro-BNP, Di-mero-D y GDF-15 fue 96,1±12,4 pg/ml, 990±140 ng/ ml y 12 ng/ml respectivamente. 15% de los pacientes (n=9) presentaban ACVs en la RNMc al momento del diagnóstico. Se observó, además, que los pacientes con ACV presentaban un SAIs más bajo que los pacientes sin eventos (5,5±1,1% y 14,6±7,3% respectivamente p=0.04). Adicionalmente, se encontró una correlación significativa entre SAIs y pro-BNP, Dimero-D y GDF-15. Conclusiones: En este trabajo se evidenció que el 15% de los pacientes con FArc presenta ACVs al momento del diagnóstico. El SAIs bajo se correlaciona de forma inversa con los biomarcadores de sobrecarga, trombogénesis, fibrosis auricular y presencia de ACV silente. Estos resultados pueden ser utilizados para una mejor estratificación del riesgo de ACV en pacientes con FA.


Introduction: Atrial myopathy and fibrosis constitute a pro-arrhythmic and pro-thromboembolic substrate in patients with atrial fibrillation (AF). Recent studies using left atrial strain (LAS) have shown that LAS contributes to predict AF recurrence in patients with paroxysmal AF. The association between LAS and cardiac biomarkers in predicting silent stroke (SS) in patients with new AF has not been studied. Aim: The association of LAS and cardiac biomarkers contribute to predict SS in patients with new AF. Methods: We have prospectively evaluated 57 consecutive patients with new AF (first episode with less than 8 weeks of evolution). Baseline clinical characteristics and blood samples for determinations of Pro-BNP, D-Dimer and GDF-15 were obtained. Brain magnetic resonance (BMRI) and 2D Echo were performed within 3 days. In sinus rhythm, the positive deflection during ventricular systole of the LAS curve derived from speckle tracking was considered (mean of 5 cycles) (LASS). Mann Whitney U test and Spearman Rho were used for statistical analysis. Results: Mean age was 70±8,2 years, 70% were men. The mean CHA2DS2-VASc score was 3,1±1. Mean pro-BNP, D-Dimer and GDF-15 were 96,1±12,4 pg/ml, 990±140 ng/ml and 12 ng/ml, respectively. Fifteen percent of patients (n=9) had evidence of previous SS in BMRI. Patients with SS had significantly less LASS than patients without events (5,5±1,1% and 14,6±7,3% respectively p=0,04). In addition, a significant correlation between LASs and pro-BNP, D-Dimer and GDF-15 was found. Conclusion: Evidence of SS was found in 15% of patients with new AF. This was associated with LASs impairment, which was inversely correlated with cardiac biomarkers of LV overload, thrombogenesis and LA fibrosis. These findings could be utilized for a better risk stratification of stroke in patients with new AF.


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Atrial Fibrillation/complications , Stroke/etiology , Peptide Fragments/blood , Prognosis , Atrial Fibrillation/diagnosis , Atrial Fibrillation/blood , Fibrin Fibrinogen Degradation Products/analysis , Magnetic Resonance Imaging , Echocardiography , Biomarkers/blood , Prospective Studies , Risk Assessment , Natriuretic Peptide, Brain/blood , Stroke/diagnosis , Stroke/blood , Growth Differentiation Factor 15/blood
11.
Rev. chil. cardiol ; 35(3): 228-241, 2016. ilus, tab
Article in Spanish | LILACS | ID: biblio-844295

ABSTRACT

La enfermedad cardiovascular se mantiene como la principal causa de morbimortalidad a nivel mundial a pesar de los avances científicos y tecnológicos recientes, por esto existe la necesidad de búsqueda de nuevas dianas terapéuticas. La autofagia es un mecanismo de degradación de proteínas y organelos disfuncionales que ocurre en vacuolas especializadas de doble membrana denominadas autofagosomas y que requiere la participación de los lisosomas. Este proceso permite el auto abastecimiento celular de energía a través del reciclaje de diversos substratos energéticos. Se activa en respuesta a diversas formas de estrés, principalmente debido a la ausencia de nutrientes y su presencia ha sido caracterizada en todos los tipos celulares que componen el sistema cardiovascular. Existe una ventana de actividad de autofagia óptima la que se relaciona con la mantención de la homeostasis cardiovascular y su desregulación participa en la patogénesis de diversas patologías cardiovasculares. En este artículo se revisa el curso temporal que llevó el descubrimiento de la autofagia, la contribución al área del Dr. Ohsumi, reciente Premio Nobel de Medicina, los principales conceptos, mecanismos celulares y moleculares de la formación del auto-fagosoma, nodos de regulación y sintetizamos su participación en la homeostasis del corazón y en la patogénesis de las enfermedades cardiovasculares y sus perspectivas futuras.


Cardiovascular disease continues to be the leading cause of morbi-mortality worldwide despite the recent scientific and technological advances. Therefore, more research is needed to discover novel therapeutic targets. Autophagy mediates the removal of dysfunctional proteins and organelles. This process takes place in double-membrane vesicles, named autophagosomes, which later fuse with lysosomes. The mechanism allows self-renewal energy repletion through diverse energy substrate recycling. Diverse forms of cellular stress, mainly nutrient deprivation, activate this process. Autophagy has been widely characterized within the cells of the cardiovascular system. There is a window of optimal autophagy activity implicated in maintaining cardiovascular homeostasis and its dysregulation participates in the pathogenesis of different cardiovascular diseases. In this article, we review the time course of auto-phagy discovery, the Nobel Prize winner Dr. Ohsumi contribution, main concepts, mechanisms involved in autophagosome formation and its regulatory no-des. Additionally, we summarized the role of auto-phagy in cardiovascular homeostasis and pathogenesis and future perspectives.


Subject(s)
Humans , Autophagy , Cardiovascular Diseases/history , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology
12.
Rev. chil. cardiol ; 35(1): 19-24, 2016. tab
Article in Spanish | LILACS | ID: lil-782638

ABSTRACT

Introducción: En pacientes con hipertensión arterial pulmonar (HAP) Galectina- 3, biomarcador de fibrosis miocárdica, se ha asociado a marcadores ecocardiográficos de remodelado ventricular derecho. La relación entre Galectina- 3, remodelado auricular derecho (AD) y capacidad funcional (CF) en pacientes con HAP no ha sido explorado. El objetivo fue medir niveles de Galectina-3 y su relación con CF y remodelado AD en pacientes con HAP Metodos: Estudio prospectivo observacional en que se incluyeron 14 pacientes con HAP En todos los pacientes se midieron los niveles de Galectina-3, proBNP, se evaluó la CF mediante test de caminata 6 minutos (TC6M) y se evaluó remodelado AD. Se consideraron para el análisis dos grupos según la distancia caminada en TC6M (> 200 m vs. ≤ 200 m). Resultados: La edad promedio fue 43 ± 10 años, el 84% mujeres. Los niveles de Galectina-3 fueron 16,1 ± 7,4 ng/mL y el TC6M fue 371 ± 142 mts. Los pacientes con TC6M< 200 m presentaron mayores niveles de Galectina-3 (27,3 ± 4,6 vs 13,7 ± 3,8; p=0,006) y mayor volumen AD (151 ± 21 vs 94 ± 43; p=0,04). Además, se observó una correlación inversa entre el área AD y TC6M (-0,71; p=0,03). Conclusión: Niveles elevados de Galectina-3 y parámetros de remodelado adverso en AD se relacionan con una menor CF en pacientes con HAP. Estos hallazgos apuntan a una mejor caracterización de pacientes con HAP y eventualmente la búsqueda de nuevos objetivos terapéuticos.


Background: Galectin-3 is a biomarker of myo-cardial fibrosis and has been associated with echocar-diographic markers of right ventricular remodeling in patients with pulmonary artery hypertension (PAH). The association among Galectin-3 level, right atrial (RA) remodeling and functional capacity (FC) has not been explored. The objective was to measure plasma Galectin-3 concentrations and its relation with RA remodeling and FC in PAH patients. Methods: This is a prospective observational study and 14 PAH patients were included. Galectin-3 and proBNP levels were measured in all patients. FC was estimated by the 6-minute walk test (6MWT) and used to define 2 groups of subjects (≤200m or >200m). RA area and volume were measured by echocardiography from a 4 chamber view. Results: The average age was 43±10 years, 84% of patients were female. Galectin-3 levels were 16.1±7.4 ng / mL and 6MWT was 371±142 m. We observed an inverse correlation between RA area and 6MWT (-0.71;p=0.03). Conclusions: Higher Galectin-3 concentrations and RA adverse remodeling are related to a decreased FC in PAH patients. These findings may lead to a better characterization of PAH patients and eventually new therapeutic targets.


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Pulmonary Artery/physiopathology , Ventricular Remodeling , Galectin 3/blood , Hypertension, Pulmonary/physiopathology , Echocardiography , Biomarkers , Prospective Studies , Observational Study , Hemodynamics , Hypertension, Pulmonary/blood
13.
Rev. chil. cardiol ; 35(1): 41-48, 2016. ilus, graf
Article in Spanish | LILACS | ID: lil-782641

ABSTRACT

El infarto del miocardio es una de las principales causas de muerte a nivel mundial y se produce a consecuencia de procesos de isquemia-reperfusión (IR). El daño miocárdico generado por IR puede ser atenuado a través del pre-condicionamiento isquémico (PI) temprano, mediado por la vía RISK o PI tardío, que se asocia a una respuesta genómica en la que se activan proteínas como óxido nítrico sintasa inducible (iNOS). Las vías de señalización que median el PI también pueden ser activadas farmacológicamente. Dexmedetomi-dina (Dex) es un agonista alfa2-adrenérgico, que se ha descrito como un potente agente cardioprotector frente a IR. Recientemente, nuestro grupo describió que Dex requiere el endotelio y la activación de la vía óxido nítrico sintasa endotelial (eNOS)-óxido nítrico (NO) para pre-condicionar el miocardio. Sin embargo, no existen estudios que muestren la posible participación de iNOS en la protección conferida por Dex. La presente adenda tiene por objetivo evaluar si Dex activa iNOS en el corazón y en cardiomiocitos. Para esto, corazones de rata adulta fueron estimulados con Dex 10 nM y se observó que el fármaco aumentó la producción de NO medida por cuantificación de nitritos, mas no estimuló la activación de iNOS medida por Western blot. Además, Dex tampoco indujo el aumento de mRNA de iNOS en cardiomiocitos adultos. Por lo tanto, Dex genera NO independiente a iNOS durante su efecto pre-condicionante agudo. Sin embargo, se requieren más estudios que clarifiquen su papel en una posible protección a largo plazo frente a IR generada por Dex.


Myocardial infarction is one of the leading causes of death worldwide and is generated as a consequence of ischemia-reperfusion (IR). Myocardial damage inflicted by IR can be attenuate by early ische-mic pre-conditioning (IP), which is mediated by the RISK pathway or late IP, which is associated to a genomic response involving the activation of proteins such as inducible nitric oxide synthase (iNOS). The signaling pathways mediating IP can also be pharmacologically activated. Dexmedetomidine (Dex) is an alpha2-adrenergic receptor agonist, which has been described as a strong cardio protective agent against IR. Recently, our group reported that Dex requires the endothelium and the activation of the endothelial nitric oxide synthase (eNOS)-ni-tric oxide (NO) pathway to precondition the myocardium. However, there are no studies showing the involvement of iNOS in the protection elicited by Dex. The aim of this Addendum is to evaluate if Dex activates iNOS in the heart and cardiomyocytes. To test this, adult rat hearts were stimulated with Dex 10 nM and we observed that NO production measured by quantification of nitrites was increased, but Dex did not activate iNOS measured by Western blot. Moreover, Dex did not induce an increase in the mRNA levels of iNOS in adult cardiomyocytes. Therefore, Dex generates NO independent of iNOS during its early pre-conditioning effect. Nevertheless, more studies are required to clarify its role in a possible long term protection against IR generated by Dex.


Subject(s)
Animals , Rats , Reperfusion Injury/prevention & control , Nitric Oxide Synthase/drug effects , Ischemic Preconditioning, Myocardial/methods , Dexmedetomidine/administration & dosage , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Cardiotonic Agents/administration & dosage , Blotting, Western , Rats, Sprague-Dawley , Disease Models, Animal , Real-Time Polymerase Chain Reaction
14.
Rev. chil. cardiol ; 34(2): 120-129, 2015. graf, tab
Article in Spanish | LILACS | ID: lil-762613

ABSTRACT

Introducción: Angiotensina (Ang)-(1-9) posee propiedades anti-hipertensivas y efecto protector a nivel cardiovascular en ratas hipertensas. Sin embargo, se desconoce si estos efectos están asociados a un mecanismo de desbalance de sodio a nivel renal. Objetivo: Determinar si el efecto anti-hipertensivo de Ang-(1-9) está asociado a un mecanismo diurético-na-triurético. Método: Ratas macho Sprague Dawley (200 ± 10g) fueron aleatorizadas para recibir Ang II (400 ng/kgmin) vía bomba osmótica. Como control se utilizaron ratas con operación sham (n=18). Después de 2 semanas desde la instalación de bomba, las ratas Sham e hipertensas fueron randomizadas para recibir vehículo (n=10), Ang-(1-9) (602 ng/kg/min, n=17) o una co-administración de Ang-(1-9) y A779 (100 ng kg-1min-1, n=7 bloqueador del receptor MAS) por 2 semanas. Resultados: Se determinó la presión arterial sistólica (PAS), masa ventricular relativa (MVR), área y perímetro de los cardiomiocitos (AC y PC) y la fracción volumétrica de colágeno total (FVCT). Para evaluar la diuresis y natriuresis se utilizaron ratas normotensas que fueron randomizadas para recibir vehículo (n=8) o Ang-(1-9) (600 ngKg-1min-1, n=8) por 6 días. Se observó un incremento significativo(p<0.05) de PAS (33%), MVR (17%), AC (64%), PC (20%), FVCT (46%). La administración crónica de Ang-(1-9) disminuyó PAS (20%), MVR (13 %), AC (35%), PC (20%) y FVCT (20%). Estos efectos no fueron mediados por el receptor MAS. Al comparar las ratas normotensas tratadas con vehículo o Ang-(1-9), se observó un aumento significativo de la diuresis y natriuresis en los días 2 y 3 en los animales con infusión de Ang-(1-9). Conclusión: Ang-(1-9) reduce la hipertensión y el remodelamiento cardíaco en ratas hipertensas. En animales normotensos se demostró que el tratamiento con Ang-(1-9)-induce diuresis y natriuresis. Este es el primer reporte que señala que el efecto de Ang-(1-9) está asociado a una regulación del sodio a nivel renal.


Background: Angiotensin-(1-9) has anti-hypertensive properties and protective cardiovascular effect in hypertensive rats. However, it is unknown whether its effects are related to a kidney mechanism to balance sodium. Aim: To determine if the anti-hypertensive effect of Ang-(1-9) is associated to a diuretic-natriuretic mechanism. Method: Sprague Dawley male rats (200±10 grs) were randomized to receive Angiotensin II by osmotic pump (400 ng/kg/min). Sham operated rats were utilized as control (n=18). Two weeks after pump setting, Sham rats with hypertension were randomized to receive placebo (n=10), Ang-(1-9)(602 ng/kg/min, n=17) or Ang-(1-9) plus A779 (Ang-(1-7) Receptor Mas blocker, 100ng/kg-1min-1, n=7) co-administration for two weeks. Arterial systolic pressure (PAS), ventricular relative mass (MVR), cardiomyocytes area and perimeter (AC and PC) and total collagen volume fraction (FVCT) were measured. Normotensive rats were utilized to evaluate diuresis and natriuresis which were randomized to receive placebo (n=8) or Ang-(1-9) (600ng/kg-1/min-1, n=8) for six days. Results: It was observed a significant rise (p<0.05) of PAS (33%), MVR (17%), AC (64%), PC (26%), FVCT (46%) was observed. Chronic administration of Ang-(1-9) reduced PAS (20%), MVR (13%), AC (35%), PC (20%) and FCVT (20%). All those effects were not mediated by Mas receptor. A significant raise was observed of diuresis and natriuresis at the second and third day of treatment in rats receiving Ang-(1-9) in comparison with normotensive rats treated with placebo. Conclusion: Ang-(1-9) reduces hypertension and cardiac remodeling in hypertensive rats. Ang-(1-9) induces natriuresis and diuresis in normotensive rats. This is the first report showing that Ang-(1-9) is associated to sodium balance in the kidney.


Subject(s)
Animals , Rats , Angiotensin II/pharmacology , Diuresis/drug effects , Natriuresis/drug effects , Antihypertensive Agents/pharmacology , Rats, Sprague-Dawley , Heart/drug effects
15.
Rev. chil. cardiol ; 31(2): 118-128, 2012. ilus, tab
Article in Spanish | LILACS | ID: lil-653792

ABSTRACT

Introducción: El polimorfismo del gen de la enzima convertidora de angiotensina I (ECA) determina mayor actividad de la ECA y mayores niveles de angioten-sina (Ang) II. Un polimorfismo similar ha sido descrito en humanos. La ECA2, a través de Ang-(1-9) más que Ang-(1-7), contrarresta los efectos deletéreos de Ang II. Se desconoce si el polimorfismo de la ECA frente a un estímulo hipertensivo modifica el eje ECA2/Ang-(1-9) y determina mayor remodelamiento de la pared aórtica de ratas hipertensas. Objetivo: Determinar el efecto del polimorfismo del gen de la ECA en la actividad del eje ECA2/Ang-(1-9) y su efecto en el remodelamiento de la pared aórtica secundaria a la hipertensión arterial (HTA) experimental. Métodos: Se usaron ratas macho homocigotas de 150 gr BN y LL. Se indujo HTA por el procedimiento Goldblatt (GB, 2 K-1clip). Ratas pseudo-operadas se usaron como controles (Sham). A las 6 semanas post cirugía se determinaron en la aorta las actividades de ECA y ECA2, los niveles de Ang II/Ang-(1-9), colágeno tipo I, células positivas para el marcador de inflamación ED-1, área y grosor de la túnica media (ATM, GTM). Resultados: El polimorfismo de la ECA con mayores niveles de ECA y Ang II determinó una mayor disminución de la actividad de ECA2, menores niveles de Ang-(1-9) y mayor remodelamiento de la pared aórtica tanto en animales normotensos como hipertensos. Conclusión: El polimorfismo de la ECA con mayor actividad de ECA y AngII determina una interregu-lación de los ejes ECA/AngII y ECA2/Ang-(1-9) lo que se asocia a mayor remodelamiento de la pared aórtica. Fondecyt 1100874.


background: The angiotensin converting enzyme (ACE) gene polymorphism determines increased ACE activity and angiotensin (Ang) II levels in Brown Norway rats (BN), compared to Lewis rats (LL). Similar polymorphism has been described in humans. ACE2 through Ang-(1-9) rather than Ang-(1-7) counteracts the deleterious effects of Ang II. It is unknown whether the ACE polymorphism counteracts the ECA2/Ang1-9 axis and determines increased remodeling of the aortic wall in hypertensive rats. Objective: To determine the effects of ACE gene polymorphism in the ECA2/Ang1-9 axis activity and its impact on the aortic wall remodeling secondary to hypertension (HT). Methods: Male homozygous rats BN and LL were used. Hypertension was induced by the Goldblatt procedure (GB, 2 K-1clip). Pseudo-operated rats were used as controls (Sham). At 6 weeks after surgery, we determined the body weight (BW) and systolic blood pressure (SBP). In aorta, we determined the ACE and ACE2 activities, Ang II/Ang1-9 levels, protein expression of collagen type I, positive cells for ED-1 inflammatory cells and medial thickness (MT) and area (MA) of aortic wall. Results: ACE polymorphism with higher levels of ACE and Ang II determined a significant decrease of ACE2 activity, Ang-(1-9) levels and aortic wall remodeling in normotensives and hypertensives rats. Conclusion: ACE polymorphism with increased ACE activity and AngII levels determines a significant inter-regulation between ACE/AngII and ACE2/Ang-(1-9) axis which is associated with increased remodeling of the aortic wall. Fondecyt 1100874.


Subject(s)
Rats , Angiotensin II/genetics , Aorta/pathology , Hypertension/genetics , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Rats, Inbred BN
16.
Rev. chil. cardiol ; 31(3): 202-214, 2012. ilus, tab
Article in Spanish | LILACS | ID: lil-670191

ABSTRACT

Resumen: La enzima convertidora de angiotensina I (ECA2) a través de Angiotensina (Ang)-(1-9) más que Ang-(1-7) contrarresta los efectos deletéreos de ECA y Ang II. Se desconoce si Ang-(1-9) es efectiva en el tratamiento del remodelamiento cardiovascular (RMCV) hipertensivo, en ratas con polimorfismo del gen de la ECA. Objetivo: Determinar el efecto de Ang-(1-9) en el tratamiento del RMCV hipertensivo en ratas con niveles genéticamente determinados de ECA y Ang II. Métodos: Ratas normotensas homocigotas, Lewis (LL) y Brown Norway (BN), se les indujo HTA a través del modelo Goldblatt (GB, 2 riñones-1 pinzado). Después de 4 semanas, las ratas hipertensas se rando-mizaron para recibir Ang-(1-9) (602 ng/Kg min) o una coadministración de Ang-(1-9)+A779 (100 ng/Kg min, antagonista del receptor MAS de Ang-(1-7)) durante 14 días mediante una minibomba. Como controles se usaron ratas sometidas a operación ficticia (Sham). Se determinó masa corporal (MC), presión arterial sistólica (PAS), masa ventricular (MV), área de cardiomiocitos (AC), área y grosor de la túnica media (ATM, GTM), fracción volumétrica de colágeno total (FVCT) en el ventrículo izquierdo (VI), niveles proteicos de colágeno tipo I (Col I) en la aorta (Ao) y la infiltración de macrófagos en Ao y VI, por medio de su molécula especifica ED1 (ED1-Ao, ED1-VI). Resultados: La administración de Ang-(1-9) disminuyó significativamente PAS, MV, AC, FVCT, Col I, ATM, GTM, ED1-Ao (-) y ED1-VI, en las ratas hipertensas LL y BN respecto a las ratas GB sin tratamiento, respectivamente. Este efecto no fue inhibido por el antagonista A779. El polimorfismo de la ECA no modificó la respuesta al tratamiento. Conclusión: Ang-(1-9) redujo eficazmente la HTA y el RMCV secundario, independiente al polimorfismo en el gen de la ECA. Este efecto posiblemente es directo ya que no fue mediado por Ang-(1-7). Fondecyt 1100874.


Background: The angiotensin I converting enzyme 2 (ACE2) counteracts the deleterious effects of ACE and Ang II through angiotensin (Ang) -(1-9) rather than Ang-(1-7). In addition, it is not clear whether Ang-(1-9) is effective in the reversal of hypertensive cardiovascular remodeling (CVRM) in rats with ACE gene polymorphism. Objective: To determine the effect of Ang-(1-9) in the prevention of hypertensive CVRM in rats with genetically determined levels of ACE and Ang II. Methods: In normotensive homozygous Lewis (LL) and Brown Norway (BN) rats hypertension was induced by the Goldblatt 2 kidney-1 pinch model. After 4 weeks, rats were randomized to receive Ang- (1-9) (602 ng / Kg min) or the co administration of Ang- (19) + A779 (100 ng / kg min, a MAS receptor antagonist of Ang- (1-7)) for 14 days. Sham operated rats were used as controls. We determined body mass (BM), systolic blood pressure (SBP), ventricular mass (VM), cardiomyocyte area (CA), area and thickness of the aortic media (ATM, TTM), LV total collagen volume fraction (FVCT), type I collagen protein levels (Col I) in the aorta (Ao) and macrophage infiltration in LV and Ao, through its specific molecule ED1 (ED1-Ao, ED1-VI). Results: Continuous administration of Ang- (1-9) significantly decreased SBP, VM, CA, TCVF, Col I, TTM, and ED1 in the aorta and left ventricle of hypertensive rats. This effect was not inhibited by the antagonist A779. ACE polymorphism did not modify the response to treatment. Conclusion: Ang- (1-9) effectively reduced hypertension induced CVRM independent of ACE gene polymorphism. This effect was not mediated by Ang- (1-7).


Subject(s)
Animals , Rats , Hypertension/chemically induced , Peptidyl-Dipeptidase A/administration & dosage , Peptidyl-Dipeptidase A/genetics , Cardiovascular System/pathology , Polymorphism, Genetic , Cardiovascular System/enzymology
17.
Rev. méd. Chile ; 139(4): 505-515, abr. 2011. ilus
Article in Spanish | LILACS | ID: lil-597648

ABSTRACT

Complications and mortality of heart failure are high, despite the availability of several forms of treatment. Uric acid, the end product of purine metabolism would actively participate in the pathophysiology of heart failure. However, there is no consensus about its action in cardiovascular disease. Serum uric acid would have a protective antioxidant activity. This action could help to reduce or counteract the processes that cause or appear as a result of heart failure. However, these protective properties would vanish in the intracellular environment or in highly hydrophobic areas such as atherosclerotic plaques and adipose tissue. This review discusses the paradoxical action of uric acid in the pathophysiology of heart failure.


Subject(s)
Animals , Humans , Heart Failure/blood , Oxidative Stress/physiology , Uric Acid/blood , Xanthine Oxidase/physiology , Biomarkers/blood , Chronic Disease , Heart Failure/physiopathology
18.
Rev. chil. cardiol ; 30(1): 34-41, 2011.
Article in Spanish | LILACS | ID: lil-592039

ABSTRACT

Introducción: La vía intracellular de RhoA/Rho kinasa es activada por agonistas de receptores acoplados a proteínas G pequeñas unidas a membrana. Su activación está relacionada al remodelado cardiovascular patológico. Previamente hemos observado aumento de actividad de Rho kinasa (ROCK) en pacientes con hipertensión arterial (HT) e hipertrofia ventricular izquierda como daño de órgano blanco. Pero su activación en relación a la diabetes no ha sido explorada en estos pacientes. Objetivo: Evaluar activación de Rho kinasa y parámetros de estrés oxidativo en pacientes hipertensos con diabetes tipo II (DMII). Métodos: Estudio comparativo entre pacientes con HT sin tratamiento, HT con DMII y hemoglobina glicosi-lada Alc > 7,5 por ciento y un grupo control normotenso. Se realizó ecocardiograma de superficie. Se midió activación de ROCK en leucocitos circulantes midiendo MYPT1 fosforilado/total (p/t) por Western blot y la velocidad de pulso carotídeo-femoral (PWV) para estimar distensibilidad arterial. El stress oxidativo se estimó midiendo ma-londialdehído (MDA) y 8-isoprostano (8-ISO) en suero. Resultados: Se incluyeron 21 pacientes hipertensos con DMII, 38 pacientes hipertensos sin DMII y 34 controles normotensos. La edad promedio fue 51 +/- 0,9; 48 +/- 0,9 y 52 (p: NS) +/- 1,1 y el 47 por ciento, 50 por ciento y 52 por ciento (p: NS) eran mujeres respectivamente. Los pacientes HT con DMII presentaron MYPTl p/t (5,6 +/- 1,3; 3,6 +/- 0,4; 2,1 +/- 0,1 p< 0,01), MDA (1,8 +/- 0,4/

Background: Rho/Rho-kinase intracellular pathway is activated by membrane bound small G-proteins. Activation of Rho/Rho-kinase pathway is related to pathologic cardiac remodeling. We have previously observed this activation (ROCK) in hypertensive patients with left ventricular hypertrophy. The influence of diabetes in this relationship has not been explored. Aim: to evaluate the activation of Rho-kinase and oxi-dative stress in hypertensive patients with type II diabetes (DMII). Methods: A comparative study between patients with untreated hypertension (HT), hypertensive patients with diabetes and hemoglobin A1c > 7.5 percent and normotensi-ve control subjects was performed. LVH was assessed by echocardiography. ROCK activity was measured in peripheral leukocytes by Western blot determination of phosphorilated / total MYPT1 ratio. Arterial compliance was determined by the relationship of carotid and femoral velocity signals (PWV) Oxidative stress was estimated by serum levels of malondialdehyde (MDA) and 8-isoprostane (8-ISO). Results: Hypertensives with DMII (n=21) had a mean age of 51 +/- 0.9 years, and 47 percent were females. Corresponding figures for 38 hypertensive patients without DM and 34 control patients were 48 ± 0,9 and 52 +/- 1,1 (NS) and 50 percent and 52 percent females, respectively (NS). The MYPT1 p/t ratio was 5,6 +/- 1,3; 3,6 +/- 0,4; 2,1 +/- 0,1 (p<0.01) in the 3 groups, respectively. MDA for the 3 groups was 1,8 +/- 0,4/

Subject(s)
Humans , Male , Adult , Female , Middle Aged , /enzymology , Hypertension/enzymology , Oxidative Stress , rho-Associated Kinases/metabolism , Analysis of Variance , Arterial Pressure , Arteriosclerosis , Blotting, Western , Cross-Sectional Studies , Echocardiography , Enzyme Activation , Isoprostanes/analysis , Malondialdehyde/analysis , Stroke Volume
19.
Rev. méd. Chile ; 138(12): 1475-1479, dic. 2010. ilus, tab
Article in Spanish | LILACS | ID: lil-583042

ABSTRACT

Background: Heart failure (HF) is characterized, among other features, by the development of alterations in myocardial energy metabolism, involving a decrease in glucose utilization and increased free fatty acid uptake by cardiomyocytes, associated with decreased deposits of high-energy phosphates (creatine phosphate/ creatine transporter). Magnetic resonance (MR) imaging allows a direct and noninvasive assessment of myocardial metabolites. Aim: To measure myocardial creatine and lipids by MR spectroscopy among patients with HF. Material and Methods: Cardiac MR spectroscopy (1.5 Tesla) with Hydrogen antenna and single voxel acquisition was performed in fve patients with non-ischemic heart failure, aged 58 ± 9.7 years, (60 percent males) and 5 healthy volunteers matched for age and sex. We analyzed the signals of creatine (Cr), lipids (L) and water (W) in the interventricular septum, establishing the water/lipid (W/L) and water/creatine (W/Cr) index to normalize the values obtained. Results: Among patients, left ventricular ejection fraction was 32 ± 6.9 percent, 60 percent were in functional capacity II, 60 percent had hypertension and one was diabetic. Spectroscopic curves showed a depletion of total Cr, evidenced by the W/ Cr index, among patients with heart failure, when compared with healthy controls (1.46 ± 1.21 and 5.96 ± 2.25 respectively, p < 0,05). Differences in myocardial lipid content, measured as the W/L index, were not significant (5.06 ± 2.66 and 1.80 ± 1.62 respectively, p = 0.08). Conclusions: Among patients with heart failure of non-ischemic etiology, there is a depletion of creatine levels measured by MR spectroscopy.


Subject(s)
Female , Humans , Male , Middle Aged , Creatine/analysis , Heart Failure/metabolism , Lipids/analysis , Magnetic Resonance Spectroscopy , Myocardium/chemistry , Case-Control Studies , Heart Failure/physiopathology , Stroke Volume/physiology , Water/chemistry
20.
Rev. méd. Chile ; 138(8): 1028-1039, ago. 2010. ilus
Article in Spanish | LILACS | ID: lil-567617

ABSTRACT

It is unknown why heart failure progresses even when patients are treated with the best therapy available. Evidences suggest that heart failure progression is due to loss of neurohumoral blockade in advanced stages of the disease and to alterations in myocardial metabolism induced, in part, by this neurohumoral activation. Alterations in cardiac energy metabolism, especially those related to substrate utilization and insulin resistance, reduce the efficiency of energy production, causing a heart energy reserve deficit. These events play a basic role in heart failure progression. Therefore, modulation of cardiac metabolism has arisen as a promissory therapy in the treatment of heart failure. This review describes myocardial energy metabolism, evaluates the role of impaired energy metabolism in heart failure progression and describes new therapies for heart failure involving metabolic intervention.


Subject(s)
Humans , Disease Progression , Energy Metabolism/physiology , Heart Failure/drug therapy , Heart Failure/physiopathology , Myocardium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL