Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Clin Sci (Lond) ; 136(21): 1537-1554, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36285636

RESUMEN

Cardiovascular diseases are the main cause of death worldwide. Recent studies have revealed the influence of histone-modifying enzymes in cardiac remodeling and heart dysfunction. The Set7 methyltransferase regulates the expression of several genes through the methylation of histones and modulates the activity of non-histone proteins. However, the role of Set7 in cardiac remodeling and heart dysfunction remains unknown. To address this question, wild-type (WT) and Set7 knockout (KO) male mice were injected with isoproterenol or saline. WT mice injected with isoproterenol displayed a decrease in Set7 activity in the heart. In addition, WT and Set7 KO mice injected with isoproterenol exhibited cardiac hypertrophy. Interestingly, Set7 deletion exacerbated cardiac hypertrophy in response to isoproterenol but attenuated myocardial fibrosis. Echocardiograms revealed that WT mice injected with isoproterenol had lowered ejection fractions and fractional shortening, and increased E'-wave deceleration time and E/A ratio compared with their controls. Conversely, Set7 KO mice did not show alteration in these parameters in response to isoproterenol. However, prolonged exposure to isoproterenol induced cardiac dysfunction both in WT and Set7 KO mice. Both isoproterenol and Set7 deletion changed the transcriptional profile of the heart. Moreover, Set7 deletion increased the expression of Pgc1α and mitochondrial DNA content in the heart, and reduced the expression of cellular senescence and inflammation markers in response to isoproterenol. Taken together, our data suggest that Set7 deletion attenuates isoproterenol-induced myocardial fibrosis and delays heart dysfunction, suggesting that Set7 plays an important role in cardiac remodeling and dysfunction in response to stress.


Asunto(s)
Cardiomiopatías , Remodelación Ventricular , Ratones , Masculino , Animales , Isoproterenol/efectos adversos , Isoproterenol/metabolismo , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Ratones Noqueados , Cardiomiopatías/inducido químicamente , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/genética , Fibrosis , Miocitos Cardíacos/metabolismo , Ratones Endogámicos C57BL
2.
Cell Death Dis ; 13(2): 144, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35145061

RESUMEN

Malaria is an enormous burden on global health that caused 409,000 deaths in 2019. Severe malaria can manifest in the lungs, an illness known as acute respiratory distress syndrome (ARDS). Not much is known about the development of malaria-associated ARDS (MA-ARDS), especially regarding cell death in the lungs. We had previously established a murine model that mimics various human ARDS aspects, such as pulmonary edema, hemorrhages, pleural effusion, and hypoxemia, using DBA/2 mice infected with Plasmodium berghei ANKA. Here, we explored the mechanisms and the involvement of apoptosis in this syndrome. We found that apoptosis contributes to the pathogenesis of MA-ARDS, primarily as facilitators of the alveolar-capillary barrier breakdown. The protection of pulmonary endothelium by inhibiting caspase activation could be a promising therapeutic strategy to prevent the pathogenicity of MA-ARDS. Therefore, intervention in the programmed death cell mechanism could help patients not to develop severe malaria.


Asunto(s)
Malaria , Síndrome de Dificultad Respiratoria , Animales , Caspasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , Malaria/complicaciones , Malaria/metabolismo , Ratones , Ratones Endogámicos DBA
3.
Bioconjug Chem ; 31(11): 2638-2647, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33169610

RESUMEN

The success of targeted drug delivery systems still requires a detailed understanding about the biological consequences of self-developed biomolecular coronas around them, since this is the surface that interacts with living cells. Herein, we report the behavior of carbohydrate-decorated amphiphilic nanoparticles in a plasma environment with regard to the formation and biological consequences of the protein corona. Naked amphiphilic nanoparticles were produced through the self-assembly of azido-PEO900-docosanoate molecules, and the coupling of N-acetylglucosamine via click chemistry enabled the fabrication of the corresponding bioactive glyco-nanostructures. Light scattering measurements, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, liquid chromatography-mass spectrometry, and the Pierce BCA protein assay all confirmed the presence of protein coronas around the self-assembled nanoparticles, regardless of the presence of the sugar residues, although it reduces the amount of adsorbed proteins. The protein coronas were formed mainly by human serum albumin, complement proteins, apolipoproteins, immunoglobulins, and proteins involved in the coagulation cascade (fibrinogen and prothrombin). While the presence of these protein coronas significantly reduced cellular uptake of the amphiphilic assemblies, they also notably reduced the cytotoxic and hemolytic effects that result from the contact of the nanoparticles with living cells. Accordingly, we highlight that protein coronas should not always be treated as artifacts that have to be avoided because they can also provide beneficial effects.


Asunto(s)
Nanopartículas/química , Corona de Proteínas/química , Adsorción , Cromatografía Liquida/métodos , Electroforesis en Gel de Poliacrilamida , Células HeLa , Humanos , Espectrometría de Masas/métodos , Microscopía Electrónica de Transmisión
4.
Cell Physiol Biochem ; 53(4): 587-605, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31535830

RESUMEN

BACKGROUND/AIMS: To investigate the role of the sympathetic nervous system (SNS) and renin-angiotensin system (RAS) in renal ischemia/reperfusion-induced (I/R) cardiac inflammatoryprofile. METHODS: Left kidney ischemia was induced in male C57BL/6 mice for 60 min, followed by reperfusion for 12 days, and treatment with or without atenolol, losartan, or enalapril. The expression of vimentin in kidney and atrial natriuretic factor (ANF) in the heart has been investigated by RT-PCR. In cardiac tissue, levels of ß1-adrenoreceptors, adenylyl cyclase, cyclic AMP-dependent protein kinase (PKA), noradrenaline, adrenaline (components of SNS), type 1 angiotensin II receptors (AT1R), angiotensinogen/Ang II and renin (components of RAS) have been measured by Western blotting and HPLC analysis. A panel of cytokines - tumour necrosis factor (TNF-α), interleukin IL-6, and interferon gamma (IFN-γ) - was selected as cardiac inflammatory markers. RESULTS: Renal vimentin mRNA levels increased by >10 times in I/R mice, indicative of kidney injury. ANF, a marker of cardiac lesion, increased after renal I/R, the values being restored to the level of Sham group after atenolol or enalapril treatment. The cardiac inflammatory profile was confirmed by the marked increase in the levels of mRNAs of TNF-α, IL-6, and IFN-γ. Atenolol and losartan reversed the upregulation of TNF-α expression, whereas enalapril restored IL-6 levels to Sham levels; both atenolol and enalapril normalized IFN-γ levels. I/R mice showed upregulation of ß1-adrenoreceptors, adenylyl cyclase, PKA and noradrenaline. Renal I/R increased cardiac levels of AT1R, which decreased after losartan or enalapril treatment. Renin expression also increased, with the upregulation returning to Sham levels after treatment with SNS and RAS blockers. Angiotensinogen/Ang II levels in heart were unaffected by renal I/R, but they were significantly decreased after treatment with losartan and enalapril, whereas increase in renin levels decreased. CONCLUSION: Renal I/R-induced cardiac inflammatory events provoked by the simultaneous upregulation of SNS and RAS in the heart, possibly underpin the mechanism involved in the development of cardiorenal syndrome.


Asunto(s)
Riñón/metabolismo , Miocardio/metabolismo , Sistema Renina-Angiotensina , Sistema Nervioso Simpático/metabolismo , Animales , Atenolol/farmacología , Atenolol/uso terapéutico , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Catecolaminas/metabolismo , Enalapril/farmacología , Enalapril/uso terapéutico , Interleucina-6/metabolismo , Losartán/farmacología , Losartán/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Sistema Nervioso Simpático/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Vimentina/genética , Vimentina/metabolismo
5.
PLoS One ; 14(4): e0214669, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30934005

RESUMEN

The sarcolemmal membrane associated proteins (SLMAPs) belong to the super family of tail anchored membrane proteins which serve diverse roles in biology including cell growth, protein trafficking and ion channel regulation. Mutations in human SLMAP have been linked to Brugada syndrome with putative deficits in trafficking of the sodium channel (Nav1.5) to the cell membrane resulting in aberrant electrical activity and heart function. Three main SLMAP isoforms (SLMAP1 (35 kDa), SLMAP2 (45 kDa), and SLMAP3 (91 kDa)) are expressed in myocardium but their precise role remains to be defined. Here we generated transgenic (Tg) mice with cardiac-specific expression of the SLMAP3 isoform during postnatal development which present with a significant decrease (20%) in fractional shortening and (11%) in cardiac output at 5 weeks of age. There was a lack of any notable cardiac remodeling (hypertrophy, fibrosis or fetal gene activation) in Tg hearts but the electrocardiogram indicated a significant increase (14%) in the PR interval and a decrease (43%) in the R amplitude. Western blot analysis indicated a selective and significant decrease (55%) in protein levels of Nav1.5 while 45% drop in its transcript levels were detectable by qRT-PCR. Significant decreases in the protein and transcript levels of the calcium transport system of the sarcoplasmic reticulum (SERCA2a/PLN) were also evident in Tg hearts. These data reveal a novel role for SLMAP3 in the selective regulation of important ion transport proteins at the level of gene expression and suggest that it may be a unique target in cardiovascular function and disease.


Asunto(s)
Corazón/fisiología , Proteínas de la Membrana/fisiología , Miocardio/metabolismo , Animales , Femenino , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica/genética , Contracción Miocárdica/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
6.
Physiol Rep ; 5(9)2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28483861

RESUMEN

C57Bl/6J mice are the gold standard animal model of diet-induced obesity. These animals become obese with higher adiposity, blood fasting glucose, triglycerides, and total cholesterol when fed a high-fat diet (HFD). Conversely, the FVB/N mouse line is thought to be resistant to diet-induced obesity, with low or no weight gain and adiposity in response to a HFD In this study, we investigated whether FVB/N mice are resistant or susceptible to metabolic disorder that is promoted by a HFD Biometric parameters and blood chemistry were analyzed in C57Bl/6J and FVB/N mice that were fed a chow diet or HFD Glucose and insulin sensitivity were assessed by performing the glucose tolerance test and measuring serum insulin/glucose and homeostasis model assessment-insulin resistance. Metabolism-related gene expression was investigated by real-time reverse transcription polymerase chain reaction. Adipocyte morphology and liver steatosis were evaluated using standard histology. FVB/N mice had higher adiposity than C57Bl/6J mice that were fed a chow diet and were glucose intolerant. FVB/N mice that were fed a HFD presented higher insulin resistance and greater liver steatosis. Epididymal white adipose tissue exhibited severe inflammation in FVB/N mice that were fed a HFD The FVB/N mouse strain is suitable for studies of diet-induced obesity, and the apparent lack of a HFD-induced response may reveal several strain-specific events that are triggered by a HFD Further studies of the FVB/N background may shed light on the complex multifactorial symptoms of obesity and metabolic syndrome.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ratones Obesos/metabolismo , Obesidad/etiología , Adiposidad , Animales , Glucemia/metabolismo , Modelos Animales de Enfermedad , Antecedentes Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos/genética , Obesidad/genética
7.
JRSM Cardiovasc Dis ; 6: 2048004016689440, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28228941

RESUMEN

BACKGROUND: Tissue remodeling is usually dependent on the deposition of extracellular matrix that may result in tissue stiffness and impaired myocardium contraction. OBJECTIVES: We had previously demonstrated that renal ischemia/reperfusion (I/R) is able to induce development of cardiac hypertrophy in mice. Therefore, we aimed to characterize renal I/R-induced cardiac hypertrophy. DESIGN: C57BL/6 J mice were subjected to 60 minutes' unilateral renal pedicle occlusion, followed by reperfusion (I/R) for 5, 8, 12 or 15 days. Gene expression, protein abundance and morphometric analyses were performed in all time points. RESULTS: Left ventricle wall thickening was increased after eight days of reperfusion (p < 0.05). An increase in the number of heart ventricle capillaries and diameter after 12 days of reperfusion (p < 0.05) was observed; an increase in the density of capillaries starting at 5 days of reperfusion (p < 0.05) was also observed. Analyses of MMP2 protein levels showed an increase at 15 days compared to sham (p < 0.05). Moreover, TGF-ß gene expression was downregulated at 12 days as well TIMP 1 and 2 (p < 0.05). The Fourier-transform infrared spectroscopy analysis showed that collagen content was altered only in the internal section of the heart (p < 0.05); such data were supported by collagen mRNA levels. CONCLUSIONS: Renal I/R leads to impactful changes in heart morphology, accompanied by an increase in microvasculature. Although it is clear that I/R is able to induce cardiac remodeling, such morphological changes is present in only a section of the heart tissue.

8.
Ther Adv Cardiovasc Dis ; 10(4): 192-202, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26715125

RESUMEN

OBJECTIVES: Thyroid hormone (TH) promotes marked effects on the cardiovascular system, including the development of cardiac hypertrophy. Some studies have demonstrated that the renin-angiotensin system (RAS) is a key mediator of the cardiac growth in response to elevated TH levels. Although some of the main RAS components are changed in cardiac tissue on hyperthyroid state, the potential modulation of the counter regulatory components of the RAS, such as angiotensin-converting enzyme type 2 (ACE2), angiotensin 1-7 (Ang 1-7) levels and Mas receptor induced by hyperthyroidism is unknown. The aim of this study was to investigate the effect of hyperthyroidism on cardiac Ang 1-7, ACE2 and Mas receptor levels. METHODS: Hyperthyroidism was induced in Wistar rats by daily intraperitoneal injections of T4 for 14 days. RESULTS: Although plasma Ang 1-7 levels were unchanged by hyperthyroidism, cardiac Ang 1-7 levels were increased in TH-induced cardiac hypertrophy. ACE2 enzymatic activity was significantly increased in hearts from hyperthyroid animals, which may be contributing to the higher Ang 1-7 levels observed in the T4 group. Furthermore, elevated cardiac levels of Ang 1-7 levels were accompanied by increased Mas receptor protein levels. CONCLUSION: The counter-regulatory components of the RAS are activated in hyperthyroidism and may be contributing to modulate the cardiac hypertrophy in response to TH.


Asunto(s)
Angiotensina I/análisis , Cardiomegalia/etiología , Hipertiroidismo/complicaciones , Miocardio/metabolismo , Fragmentos de Péptidos/análisis , Peptidil-Dipeptidasa A/análisis , Proteínas Proto-Oncogénicas/análisis , Receptores Acoplados a Proteínas G/análisis , Angiotensina I/fisiología , Enzima Convertidora de Angiotensina 2 , Animales , Masculino , Fragmentos de Péptidos/fisiología , Peptidil-Dipeptidasa A/fisiología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/fisiología , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/fisiología , Sistema Renina-Angiotensina/fisiología
9.
Int J Cardiol ; 171(2): 199-208, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24374203

RESUMEN

OBJECTIVES: In the present study, we investigated whether MSC-transplantation can revert cardiac dysfunction in streptozotocin-induced diabetic rats and the immunoregulatory effects of MSC were examined. BACKGROUND: Cardiac complications are one of the main causes of death in diabetes. Several studies have shown anti-diabetic effects of bone marrow mesenchymal stromal cells (MSC). METHODS/RESULTS: The rats were divided in three groups: Non-diabetic, Diabetic and Diabetic-Treated with 5 × 10(6) MSC 4 weeks after establishment of diabetes. Four weeks after MSC-therapy, systemic metabolic parameters, immunological profile and cardiac function were assessed. MSC-transplantation was able to revert the hyperglycemia and body weight loss of the animals. In addition, after MSC-transplantation a decrease in corticosterone and IFN-γ sera levels without restoration of insulin and leptin plasma levels was observed. Also, MSC-therapy improved electrical remodeling, shortening QT and QTc in the ECG and action potential duration of left ventricular myocytes. No arrhythmic events were observed after MSC-transplantation. MSC-therapy rescued the cardiac beta-adrenergic sensitivity by increasing beta-1 adrenergic receptor expression. Both alpha and beta cardiac AMPK and p-AMPK returned to baseline values after MSC-therapy. However, total ERK1 and p-ERK1/2 were not different among groups. CONCLUSION: The results indicate that MSC-therapy was able to rescue cardiac impairment induced by diabetes, normalize cardiac AMPK subunit expression and activity, decrease corticosterone and glycemia and exert systemic immunoregulation.


Asunto(s)
Complicaciones de la Diabetes/terapia , Diabetes Mellitus Experimental/complicaciones , Cardiopatías/terapia , Hiperglucemia/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Corticosterona/sangre , Complicaciones de la Diabetes/etiología , Complicaciones de la Diabetes/inmunología , Diabetes Mellitus Experimental/inmunología , Sistema de Conducción Cardíaco/fisiología , Cardiopatías/etiología , Cardiopatías/inmunología , Hiperglucemia/etiología , Hiperglucemia/inmunología , Masculino , Células Madre Mesenquimatosas , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Remodelación Ventricular/fisiología
10.
Vascul Pharmacol ; 52(3-4): 166-70, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19857605

RESUMEN

Thyroid hormones (THs) exert multiple effects on the heart and vascular system. As a consequence, altered cardiovascular function observed in the thyroid diseases corresponds to one of the most important and clinically relevant aspects found in both hyperthyroidism and hypothyroidism. Besides THs' direct effects on the heart and vascular system, in the last three decades several studies have implicated the Renin-Angiotensin System (RAS) in some of the cardiovascular effects of THs, with this interaction suggesting that RAS may be an important mediator of THs actions. In the present review, we discuss the alterations in the circulating RAS, as well as modifications in cardiac and vascular RAS which are involved in the cardiovascular alterations found during the modulation of TH levels. In addition, considering the important role that both systems present during fetal and neonatal periods, we also review the interaction between THs and the RAS in the development of cardiovascular system. A greater understanding of the role of the RAS in hyperthyroidism and hypothyroidism, during early or adult life will presumably facilitate the evolution of newer, targeted therapies.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Sistema Renina-Angiotensina/fisiología , Hormonas Tiroideas/metabolismo , Adulto , Animales , Enfermedades Cardiovasculares/fisiopatología , Sistemas de Liberación de Medicamentos , Desarrollo Fetal , Humanos , Hipertiroidismo/complicaciones , Hipertiroidismo/fisiopatología , Hipotiroidismo/complicaciones , Hipotiroidismo/fisiopatología , Recién Nacido
11.
Clin Sci (Lond) ; 114(5): 375-80, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17953515

RESUMEN

There have been only a few reports on the sympathoadrenal and renin-angiotensin systems in children of small gestational age. The purpose of the present study was to investigate plasma levels of ACE (angiotensin-converting enzyme) activity, angiotensin and catecholamines in 8- to 13-year-old children and to determine whether there are correlations between the components of these systems with both birthweight and BP (blood pressure) levels. This clinical study included 66 children (35 boys and 31 girls) in two groups: those born at term with an appropriate birthweight [AGA (appropriate-for-gestational age) group, n=31] and those born at term but with a small birthweight for gestational age [SGA (small-for-gestational age) group, n=35]. Concentrations of angiotensin, catecholamines and ACE activity were determined in plasma. Circulating noradrenaline levels were significantly elevated in SGA girls compared with AGA girls (P=0.036). In addition, angiotensin II and ACE activity were higher in SGA boys (P=0.024 and P=0.050 respectively). There was a significant association of the circulating levels of both angiotensin II and ACE activity with BP levels in our study population. Although the underlying mechanisms that link restricted fetal growth with later cardiovascular events are not fully understood, the findings in the present study support the link between low birthweight and overactivity of both sympathoadrenal and renin-angiotensin systems into later childhood.


Asunto(s)
Peso al Nacer/fisiología , Recién Nacido Pequeño para la Edad Gestacional/fisiología , Sistema Renina-Angiotensina/fisiología , Adolescente , Angiotensina II/sangre , Presión Sanguínea/fisiología , Niño , Cromatografía Líquida de Alta Presión/métodos , Estudios Transversales , Femenino , Edad Gestacional , Humanos , Recién Nacido , Masculino , Norepinefrina/sangre , Peptidil-Dipeptidasa A/sangre
12.
Life Sci ; 80(8): 782-7, 2007 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17161436

RESUMEN

Epidemiological studies suggest that intrauterine undernutrition plays an important role in the development of arterial hypertension and endothelial dysfunction in adulthood. We have evaluated the effect of the Renin Angiotensin System inhibition on the blood pressure and the mesenteric arteriolar reactivity of the intrauterine undernourished rats. Wistar rats were fed either normal or 50% of the normal intake diets, during the whole gestational period. In this study only the male offspring was used. At 16 weeks of age, the rats were used for the study of blood pressure, microvascular reactivity studied in vivo-in situ to Angiotensin II (Ang II), Bradykinin (Bk) and Acetylcholine (Ach) before and after either losartan (10 mg/kg/15 days) or enalapril (15 mg/kg/21 days) treatment. We also evaluated the mesenteric and plasmatic Angiotensin Converting Enzyme (ACE), renal function, lipid plasmatic content, and insulin and glucose metabolism. Intrauterine undernutrition induced hypertension and increased response of mesenteric arterioles to Ang II and decreased vasodilation to Bk and Ach. The treatments with losartan or enalapril normalized the blood pressure levels and significantly improved the arteriolar responses to Bk, Ach and reduced the response to Ang II. No differences have been detected to ACE activity, renal function, lipid content and insulin and glucose metabolism. This study shows for the first time that Renin Angiotensin System inhibitors can normalize the cardiovascular alterations induced by intrauterine undernutrition.


Asunto(s)
Antihipertensivos/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Enalapril/uso terapéutico , Privación de Alimentos , Losartán/uso terapéutico , Desnutrición/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiopatología , Glucemia/análisis , Antagonismo de Drogas , Femenino , Retardo del Crecimiento Fetal/fisiopatología , Insulina , Masculino , Mesenterio/irrigación sanguínea , Embarazo , Ratas , Ratas Wistar , Sistema Renina-Angiotensina/fisiología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
13.
Virchows Arch ; 448(4): 472-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16440199

RESUMEN

Cardiac hypertrophy in response to hyperthyroidism is well known. However, the effects on cardiac microcirculation are still controversial in this model. The present study evaluated the effects of acute administration of two different thyroxine (T4) dose levels on the angiogenic response in the myocardium. Capillary density (CD), the CD to fiber density (FD) ratio (CD/FD), and intercapillary distance (ICD) were assessed, as was ventricle weight (VW) to body weight (BW) ratio (VW/BW). Collagen I and III messenger ribonucleic acid (mRNA) expression and VEGF-A expression were also determined by reverse transcriptase polymerase chain reaction (RT-PCR). Immunohistochemical detection of proliferating cell nuclear antigen (PCNA) expression in endothelial cell nuclei was also carried out. We simulated an acute hyperthyroidism situation in male Wistar rats by daily intraperitoneal injection of T4 (0.025 or 0.1 mg kg(-1) day(-1)) for 7 days. Hemodynamic parameters showed that T4 did not alter systolic blood pressure (SBP) but significantly increased heart rate (HR). Both T4 doses significantly increased VW. Morphologically, the higher T4 dose resulted in a 33% greater myocardial mass, which was not accompanied by alterations in collagen I and III mRNA expression. The CD and CD/FD parameters were significantly lower in the hyperthyroid rats treated with the higher dose than in the control animals, and PCNA-labeling analysis indicated total absence of marked capillary growth. However, although the acute treatment with T4 did not induce any alteration in capillary number and endothelial cell proliferation, the vascular endothelial growth factor (VEGF)-A mRNA and protein expression were significantly increased with the higher T4 dose. These data indicate that the cardiac hypertrophy induced by acute treatment with thyroid hormone precedes the angiogenic process, which probably occurs later.


Asunto(s)
Cardiomegalia/inducido químicamente , Corazón/efectos de los fármacos , Tiroxina/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Capilares/efectos de los fármacos , Capilares/patología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Masculino , Miocardio/patología , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/patología , Tamaño de los Órganos/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA