Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 119: 75-86, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29723491

RESUMEN

QSOX1, a sulfhydryl oxidase, was shown to be upregulated in the heart upon acute heart failure (AHF). The aim of the study was to unravel QSOX1 roles during AHF. We generated and characterized mice with QSOX1 gene deletion. The QSOX1-/- mice were viable but adult male exhibited a silent dilated cardiomyopathy. The QSOX1-/- hearts were characterized by low protein SERCA2a levels associated with a calcium homeostasis alteration, high levels of the endoplasmic reticulum (ER) chaperone proteins Grp78/Bip, and of the ER apoptosis sensor CHOP, indicating a chronic unfolded protein response (UPR). Importantly the QSOX1invalidation led to overexpression of two ER oxidases, ERO1-α and PRDX4. Acute stress was induced by isoproterenol injection (ISO, 300 mg/kg/12 h) for 2 days. In both groups, the PERK UPR pathway was transiently activated 6 h after the first ISO injection as indicated by eIF2 phosphorylation. By day-3 after the onset of stress, both WT and QSOX1-/- mice exhibited AHF profile but while high cardiac QSOX1 level was induced in WT hearts, ERO1-α and PRDX4 levels drop down in QSOX1-/-. At that time, QSOX1-/- hearts exhibited an enhanced inflammation (CD68+ cells and Galectin-3 expression) and oxidative stress (DHE staining and oxyblot) when compared to WT ones. In conclusion, the lack of QSOX1 promotes the upregulation of two ER oxidases ERO1α and PRDX4 that likely rescues oxidative protein folding in the hearts. However, signs of chronic ER stress remained present and were associated with a dilated cardiomyopathy. The superimposition of acute stress allowed us to propose that QSOX1 participate to the early response to cardiac stress but not to immediate UPR response. Taken altogether, the data indicated that QSOX1 is required 1) for a proper protein folding in the endo/sarcoplasmic reticulum (ER/SR) and 2) for resolution and protective response during acute stress.


Asunto(s)
Cardiomiopatía Dilatada/genética , Insuficiencia Cardíaca/genética , Inflamación/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Animales , Apoptosis/genética , Calcio/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Eliminación de Gen , Regulación de la Expresión Génica/genética , Glicoproteínas/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Inflamación/fisiopatología , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo/genética , Oxidorreductasas , Peroxirredoxinas/genética , Pliegue de Proteína , Retículo Sarcoplasmático , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Factor de Transcripción CHOP/genética , Respuesta de Proteína Desplegada/genética
2.
FASEB J ; 28(6): 2430-40, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24558200

RESUMEN

Studies have shown that aldosterone would have angiogenic effects and therefore would be beneficial in the context of cardiovascular diseases. We thus investigated the potential involvement of aldosterone in triggering a cardiac angiogenic response in the context of type-2 diabetes and the molecular pathways involved. Male 3-wk-old aldosterone synthase (AS)-overexpressing mice and their control wild-type (WT) littermates were fed a standard or high-fat, high-sucrose (HFHS) diet. After 6 mo of diet treatment, mice were euthanized, and cardiac samples were assayed by RT-PCR, immunoblotting, and immunohistology. HFHS diet induced type-2 diabetes in WT (WT-D) and AS (AS-D) mice. VEGFa mRNAs decreased in WT-D (-43%, P<0.05 vs. WT) and increased in AS-D mice (+236%, P< 0.01 vs. WT-D). In WT-D mouse hearts, the proapoptotic p38MAPK was activated (P<0.05 vs. WT and AS-D), whereas Akt activity decreased (-64%, P<0.05 vs. WT). The AS mice, which exhibited a cardiac up-regulation of IGF1-R, showed an increase in Akt phosphorylation when diabetes was induced (P<0.05 vs. WT and AS-D). Contrary to WT-D mice, AS-D mouse hearts did not express inflammatory markers and exhibited a normal capillary density (P<0.05 vs. WT-D). To our knowledge, this is the first study providing new insights into the mechanisms whereby aldosterone prevents diabetes-induced cardiac disorders.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Aldosterona/farmacología , Animales , Glucemia/metabolismo , Citocromo P-450 CYP11B2/biosíntesis , Citocromo P-450 CYP11B2/genética , Dieta Alta en Grasa , Corazón/efectos de los fármacos , Hiperaldosteronismo/fisiopatología , Resistencia a la Insulina , Masculino , Ratones , Miocardio/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/biosíntesis
3.
Curr Hypertens Rep ; 15(4): 395-400, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23686824

RESUMEN

Cardiac remodeling is a deleterious consequence of arterial hypertension. This remodeling results from cardiac transcriptomic changes induced by mechanical and hormonal factors. Angiotensin II and aldosterone often collaborate in pathological situations to induce hypertrophy of cardiomyocytes, vascular inflammation, perivascular and interstitial fibrosis, and microvascular rarefaction. Experimental models of transgenic mice overexpressing renin in liver, leading to increased plasma angiotensin II and severe hypertension, and mice overexpressing aldosterone-synthase in cardiomyocytes, leading to a doubling of intracardiac aldosterone concentration have shown that cardiac fibrosis in the heart depends on a balance between pro-fibrotic (TGF-ß, galectin-3) and anti-fibrotic (BNP, ANP) factors. Recent studies using cell-specific deletion of the mineralocorticoid receptor indicate that its activation in macrophages is a key step in the development of cardiac fibrosis in the setting of hemodynamic or hormonal challenges. This review focuses on the impact of inappropriate stimulation of aldosterone in the development of cardiac fibrosis.


Asunto(s)
Aldosterona/metabolismo , Cardiopatías/metabolismo , Hipertensión/metabolismo , Angiotensina II/metabolismo , Animales , Citocromo P-450 CYP11B2/metabolismo , Fibrosis/etiología , Fibrosis/metabolismo , Cardiopatías/fisiopatología , Humanos , Hipertensión/fisiopatología
4.
J Biol Chem ; 286(40): 35007-19, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21768101

RESUMEN

Alterations in the balance of cytoskeleton as well as energetic proteins are involved in the cardiac remodeling occurring in dilated cardiomyopathy (DCM). We used two-dimensional DIGE proteomics as a discovery approach to identify key molecular changes taking place in a temporally controlled model of DCM triggered by cardiomyocyte-specific serum response factor (SRF) knock-out in mice. We identified muscle creatine kinase (MCK) as the primary down-regulated protein followed by α-actin and α-tropomyosin down-regulation leading to a decrease of polymerized F-actin. The early response to these defects was an increase in the amount of desmin intermediate filaments and phosphorylation of the αB-crystallin chaperone. We found that αB-crystallin and desmin progressively lose their striated pattern and accumulate at the intercalated disk and the sarcolemma, respectively. We further show that desmin is a preferential target of advanced glycation end products (AGE) in mouse and human DCM. Inhibition of CK in cultured cardiomyocytes is sufficient to recapitulate both the actin depolymerization defect and the modification of desmin by AGE. Treatment with either cytochalasin D or glyoxal, a cellular AGE, indicated that both actin depolymerization and AGE contribute to desmin disorganization. Heat shock-induced phosphorylation of αB-crystallin provides a transient protection of desmin against glyoxal in a p38 MAPK-dependent manner. Our results show that the strong down-regulation of MCK activity contributes to F-actin instability and induces post-translational modification of αB-crystallin and desmin. Our results suggest that AGE may play an important role in DCM because they alter the organization of desmin filaments that normally support stress response and mitochondrial functions in cardiomyocytes.


Asunto(s)
Actinas/metabolismo , Cardiomiopatía Dilatada/metabolismo , Forma MM de la Creatina-Quinasa/deficiencia , Forma MM de la Creatina-Quinasa/genética , Desmina/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Alelos , Animales , Electroforesis en Gel Bidimensional , Ventrículos Cardíacos/patología , Homocigoto , Humanos , Espectrometría de Masas/métodos , Ratones , Modelos Biológicos , Miocitos Cardíacos/citología , Ratas , Tropomiosina/metabolismo , Cadena B de alfa-Cristalina/química
5.
J Cardiovasc Pharmacol ; 60(3): 315-21, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22710814

RESUMEN

BACKGROUND: Aldosterone antagonists (AAs) have beneficial effects on ventricular histological and electrical remodeling and improve noradrenaline uptake. Adding an AA to a beta-blocker (BB) further improves cardiac mortality in heart failure patients. We investigated if adjunction of a BB modifies beneficial effects of spironolactone on different parameters of the ventricular remodeling. METHODS: A severe myocardial infarction (MI) was produced in rats. Three months after surgery, left ventricular (LV) function was assessed by echocardiography. Fifty-five rats with heart failure were then randomized in 5 groups: sham, MI, and MI treated for 4 weeks with spironolactone (10 mg·kg·d), atenolol (1 mg·kg·d), or both. Holter transducers were implanted to record 24-hour ventricular electrical parameters, mean cycle length (RR) and SD of RR. Before killing, invasive left ventricular end diastolic pressure (LVEDP) was recorded. LV samples were used for histological analysis and catecholamine assay. RESULTS: Rats with MI had significantly increased LVEDP (32 ± 3 vs. 14 ± 1 mm Hg), LV, collagen content (5.8% ± 1.4% vs. 3.6% ± 0.7%), ventricular premature complexes (2.5 10 ± 10 vs. 30 ± 13), and decreased meanRR (164 ± 2 vs. 169 ± 1 milliseconds) and SDRR (3.9 ± 0.2 vs. 5.4 ± 0.2 milliseconds) compared with sham. At nonhypotensive doses, spironolactone and atenolol similarly improved LVEDP. Compared with MI, although spironolactone significantly decreased ventricular premature complexes, LV collagen and noradrenaline contents, and improved meanRR and SDRR, atenolol had effects only on meanRR and SDRR. Addition of atenolol to spironolactone further improved spironolactone effects on all these parameters. CONCLUSIONS: AA improved, independently of the cardiac function, histological and electrical remodeling after MI. A BB added to an AA did not blunt these beneficial effects; furthermore, it improved these effects related to spironolactone.


Asunto(s)
Antagonistas Adrenérgicos beta/administración & dosificación , Insuficiencia Cardíaca/patología , Miocardio/patología , Índice de Severidad de la Enfermedad , Espironolactona/administración & dosificación , Remodelación Ventricular/efectos de los fármacos , Animales , Enfermedad Crónica , Quimioterapia Combinada , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Resultado del Tratamiento , Remodelación Ventricular/fisiología
6.
Mol Cell Biochem ; 347(1-2): 63-70, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20938722

RESUMEN

Hyperhomocysteinemia, characterized by an elevated plasma homocysteine concentration, leads to several clinical manifestations and particularly cardiovascular diseases. Experimental models of hyperhomocysteinemia revealed several tissue injuries including heart fibrosis and ventricular hypertrophy. In order to analyze the molecular mechanisms link to these morphological alterations, a mild hyperhomocysteinemia was induced in rats via a chronic methionine administration. Effects of methionine administration were examined by histological analysis with Sirius red staining, histomorphometric analysis, zymography, and immunoblotting. Hyperhomocysteinemia due to methionine administration produces an interstitial myocardial fibrosis and a ventricular cardiomyocyte hypertrophy, which were associated with increased expression of transforming growth factor-beta1 (TGFß1), tissue inhibitors of metalloproteinase (TIMP) 2, and JNK activation. However, the matrix metalloproteinase 2 activity was decreased in the hearts of hyperhomocysteinemic rats. Moreover, the TIMP1 protein expression was decreased, and the TIMP1-MMP1 balance was shifted. Remodeling in cardiac tissue observed in rat model of mild hyperhomocysteinemia is associated with a dysregulation in extracellular matrix degradation which results, at least in part, from enhancement of TGFß1 level.


Asunto(s)
Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/patología , Miocardio/metabolismo , Miocardio/patología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Colágeno/metabolismo , Activación Enzimática/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Homocisteína/sangre , Homocisteína/farmacología , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Miocardio/enzimología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Wistar , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
7.
FASEB J ; 23(7): 2176-85, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19255254

RESUMEN

Recent reports showed an unexpected worsening of endothelial function by aldosterone antagonism in diabetic patients, suggesting that aldosterone could interfere with the detrimental consequences of diabetes on microvasculature and thus on cardiac function. To test this hypothesis, diabetes (D) was induced using streptozotocin in transgenic (Tg) male mice overexpressing aldosterone-synthase in the heart and in wild-type (Wt) mice. Eight weeks after streptozotocin injection, impairment of left ventricular systolic function, measured by echocardiography (fractional shortening), was accompanied by a decrease in capillaries/cardiomyocyte ratio (-20%) and VEGFa expression (-40%) in Wt-D mice compared with normoglycemic littermates. Furthermore, Wt-D mice demonstrated an increase in superoxide production (+100%) and protein carbonylation (+33%), hallmarks of oxidative stress. Except for a slight increase in protein carbonylation, all of these diabetes-associated cardiac alterations were undetectable in Tg-D mice. Fibrosis was induced similarly in both diabetic groups. Eplerenone (an aldosterone antagonist) abolished all of the effects of aldosterone-synthase overexpression but had no effect in Wt-D mice. Thus, aldosterone prevents systolic dysfunction through a mineralocorticoid receptor-dependent mechanism that may include preventing VEGFa down-regulation and maintaining capillary density. Understanding how aldosterone prevents VEGFa down-regulation in experimental diabetes could be important to define new strategies targeting the prevention of a decrease in capillary density.


Asunto(s)
Aldosterona/fisiología , Capilares , Vasos Coronarios , Regulación de la Expresión Génica/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Aldosterona/genética , Animales , Complicaciones de la Diabetes , Diabetes Mellitus Experimental , Corazón , Ratones , Miocardio , Receptores de Mineralocorticoides/fisiología , Sístole , Disfunción Ventricular Izquierda
8.
Resuscitation ; 156: 251-259, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32858156

RESUMEN

PURPOSE: Early and reliable prognostication after cardiac arrest (CA) remains crucial. We hypothesized that protein-S100B (PS100B) could predict more accurately outcome in the early phase of CA compared with other current biomarkers. METHODS: This prospective single-center study included 330 adult comatose non-traumatic successfully resuscitated CA patients, treated with targeted temperature management but not extra-corporeal life support. Lactate, pH, creatinine, NSE, and PS100B were sampled in ICU early after return of spontaneous circulation (ROSC) corresponding to admission (Adm). Serial measurements were also performed at H24 and H48. PS100B was the sole biomarker blinded to physicians. MEASUREMENTS AND MAIN RESULTS: The median delay between ROSC and first PS100B sampling was 220 min. At admission, all biomarkers were significantly associated with good outcome (CPC1-2; 109 patients) at 3-month follow-up (P ≤ 0.001, except for NSE: P = 0.03). PS100B-Adm showed the best AUC of ROC curves for outcome prediction at 3-month (AUC 0.83 [95%-CI: 0.78-0.88]), compared with other biomarkers (P < 0.0001), while AUC for lactate-Adm was higher than for NSE-Adm. AUC for PS100B-H24 was significantly higher than for other biomarkers except NSE-H24 (P ≤ 0.0001), while AUC for NSE-H24 was higher than for lactate-H24 and pH-H24. AUCs for PS100-H48 and NSE-H48 were significantly higher than for all other biomarkers (P < 0.001). Compared to patients with decreased PS100B values over time, an increasing PS100B value between admission and H24 was significantly associated with poor outcome at 3 months (P = 0.001). No-flow, initial non-shockable rhythm, PS100B-Adm, lactate-Adm, pH-Adm, clinical seizures, and absence of therapeutic hypothermia were independent predictors associated with poor outcome at 3-month in multivariate analysis. Net-Reclassification-Index was 70%, 64%, and 81% when PS100B-Adm was added to the clinical model, to clinical model with NSE-Adm, and to clinical model with standard biological parameters, respectively. CONCLUSIONS: Early PS100B compared with other biomarkers was independently correlated with outcome after CA, with an interesting added value.


Asunto(s)
Paro Cardíaco , Hipotermia Inducida , Adulto , Biomarcadores , Paro Cardíaco/terapia , Humanos , Fosfopiruvato Hidratasa , Pronóstico , Estudios Prospectivos , Curva ROC , Subunidad beta de la Proteína de Unión al Calcio S100
9.
Pharmacol Rep ; 61(1): 58-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19307693

RESUMEN

Our understanding of the effects of aldosterone and its mechanisms has increased substantially in recent years, probably because of the importance of the mineralocorticoid receptor (MR) antagonists in several major cardiovascular diseases. Recent clinical studies have confirmed the benefits of MR antagonists in patients with heart failure, left ventricular dysfunction after myocardial infarction, hypertension or diabetic nephropathy. However, it would be a gross oversimplification to conclude that the role of aldosterone is unequivocally negative. Aldosterone is synthesized in the adrenal glands and binds to specific MRs in target epithelial cells. The steroid-receptor complex penetrates the cell nucleus where it modulates gene expression and activates specific aldosterone-induced proteins that control sodium reabsorption. Recent studies have shown that aldosterone also impacts a wide range of non-epithelial tissues such as the heart and blood vessels. Remarkably, aldosterone can also be synthesized in extra-adrenal tissues and it may act in a rapid non-genomic manner.We note the existence of glucocorticoids that exhibit plasma concentrations much higher than those of aldosterone and that are structurally very similar to aldosterone. It is thus possible that glucocorticoids may bind to the aldosterone receptor in some cell types. Diverse experimental models and several strains of transgenic mice have allowed us to better understand the effects of aldosterone on the heart. Specifically, it seems that a slight increase in cardiac aldosterone concentrations induces a decreased coronary reserve in mice by decreasing the BKCa potassium channels associated with coronary smooth muscle cells. Taken together, these experiments indicate that vascular cells are the primary targets of aldosterone in the cardiovascular system. The hormone directly affects NO and EDHF-mediated coronary relaxation. Both mechanisms may contribute to the deleterious cardiovascular effects of MR stimulation.


Asunto(s)
Aldosterona/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Antagonistas de Receptores de Mineralocorticoides , Aldosterona/biosíntesis , Angiotensina II/metabolismo , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Ensayos Clínicos como Asunto , Humanos , Transducción de Señal
10.
JACC Basic Transl Sci ; 4(6): 717-732, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31709320

RESUMEN

Acute kidney injury is associated with increased risk of heart failure and mortality. This study demonstrates that acute kidney injury induces remote cardiac dysfunction, damage, injury, and fibrosis via a galectin-3 (Gal-3) dependent pathway. Gal-3 originates from bone marrow-derived immune cells. Cardiac damage could be prevented by blocking this pathway.

11.
Circulation ; 116(21): 2435-43, 2007 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-17984374

RESUMEN

BACKGROUND: Cardiomyocyte-specific overexpression of aldosterone synthase in male (MAS) mice induces a nitric oxide-independent coronary dysfunction. Because calcium-activated potassium (BKCa) channels are essential for vascular smooth muscle cell (VSMC) relaxation, we hypothesized that aldosterone alters their expression and/or function in VSMCs. METHODS AND RESULTS: Left coronary artery segments were isolated from MAS or male wild-type mice and mounted in a wire myograph. Responses to acetylcholine were assessed (in the presence of a nitric oxide synthase inhibitor) without or with the cyclooxygenase inhibitor diclofenac, the KCa inhibitors charybdotoxin plus apamin, or the BKCa inhibitor iberiotoxin. Expression of BKCa was quantified in hearts by real-time quantitative polymerase chain reaction and Western blot and in isolated coronary arteries by polymerase chain reaction. The effect of aldosterone on BKCa expression also was studied in cultured rat aortic VSMCs. Acetylcholine-mediated coronary relaxation was markedly decreased in MAS mice and was prevented by spironolactone. Diclofenac did not affect the MAS-induced impairment in the responses to acetylcholine, whereas charybdotoxin plus apamin virtually abolished the relaxation in both male wild-type and MAS mice. After iberiotoxin, relaxation to acetylcholine was decreased to a larger extent in male wild-type than in MAS, leading to similar levels of relaxation. BKCa-alpha and -beta1 subunit expressions were significantly decreased in MAS heart and coronary arteries. In cultured VSMCs, aldosterone induced a concentration-dependent decrease in BKCa expression, which was prevented by spironolactone. CONCLUSIONS: Aldosterone overexpression altered VSMC BKCa expression and coronary BKCa-dependent relaxation. The resulting alteration of relaxing responses may contribute to the deleterious effects of aldosterone in cardiovascular diseases. BKCa channels may therefore be useful therapeutic targets in cardiovascular diseases.


Asunto(s)
Aldosterona/fisiología , Enfermedad Coronaria/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canales de Potasio Calcio-Activados/fisiología , Aldosterona/biosíntesis , Aldosterona/genética , Animales , Células Cultivadas , Enfermedad Coronaria/genética , Enfermedad Coronaria/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , Canales de Potasio Calcio-Activados/genética , Vasodilatación/fisiología
12.
Cardiovasc Res ; 72(1): 69-79, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16839528

RESUMEN

OBJECTIVES: The expression and distribution of connexins is abnormal in a number of cardiac diseases, including atrial fibrillation, and is believed to favor conduction slowing and arrhythmia. Here, we studied the role of atrial structural remodeling in the disorganization of gap junctions and whether redistributed connexins can form new functional junction channels. METHODS: Expression of connexin-43 (Cx43) was characterized by immunoblotting and immunohistochemistry in human right atrial specimens and in rat atria after myocardial infarction (MI). Gap junctions were studied by electron and 3-D microscopy, and myocyte-myocyte coupling was determined by Lucifer yellow dye transfer. RESULTS: In both chronically hemodynamically overloaded human atria in sinus rhythm and in dilated atria from MI-rats, Cx43 were dephosphorylated and redistributed from the intercalated disc to the lateral cell membranes as observed during atrial fibrillation. In MI-rats, the gap junctions at the intercalated disc were smaller (20% decrease) and contained very little Cx43 (0 or 1 gold particle vs. 42 to 98 in sham-operated rats). In the lateral membranes of myocytes, numerous connexon aggregates comprising non-phosphorylated Cx43 were observed. These connexon aggregates were in no case assembled into gap junction plaque-like structures. However, N-cadherin was well organized in the intercalated disc. There was very little myocyte-myocyte coupling in MI-rat atria and no myocyte-fibroblast coupling. Regression of the atrial remodeling was associated with the normalization of Cx43 localization. CONCLUSION: Structural alteration of the atrial myocardium is an important factor in the disorganization of connexins and gap junction. Moreover, redistributed Cx43 do not form junction channels.


Asunto(s)
Fibrilación Atrial/patología , Conexina 43/análisis , Uniones Comunicantes/patología , Atrios Cardíacos/ultraestructura , Animales , Fibrilación Atrial/metabolismo , Comunicación Celular , Fibrosis , Técnica de Fractura por Congelación , Uniones Comunicantes/metabolismo , Atrios Cardíacos/metabolismo , Humanos , Imagenología Tridimensional , Immunoblotting , Inmunohistoquímica , Microscopía Electrónica , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Ratas
13.
J Appl Physiol (1985) ; 123(6): 1579-1591, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28883044

RESUMEN

Exercise training has been demonstrated to have beneficial effects in patients with heart failure (HF) or diabetes. However, it is unknown whether diabetic patients with HF will benefit from exercise training. Male Wistar rats were fed either a standard (Sham, n = 53) or high-fat, high-sucrose diet ( n = 66) for 6 mo. After 2 mo of diet, the rats were already diabetic. Rats were then randomly subjected to either myocardial infarction by coronary artery ligation (MI) or sham operation. Two months later, heart failure was documented by echocardiography and animals were randomly subjected to exercise training with treadmill for an additional 8 wk or remained sedentary. At the end, rats were euthanized and tissues were assayed by RT-PCR, immunoblotting, spectrophotometry, and immunohistology. MI induced a similar decrease in ejection fraction in diabetic and lean animals but a higher premature mortality in the diabetic group. Exercise for 8 wk resulted in a higher working power developed by MI animals with diabetes and improved glycaemia but not ejection fraction or pathological phenotype. In contrast, exercise improved the ejection fraction and increased adaptive hypertrophy after MI in the lean group. Trained diabetic rats with MI were nevertheless able to develop cardiomyocyte hypertrophy but without angiogenic responses. Exercise improved stress markers and cardiac energy metabolism in lean but not diabetic-MI rats. Hence, following HF, the benefits of exercise training on cardiac function are blunted in diabetic animals. In conclusion, exercise training only improved the myocardial profile of infarcted lean rats fed the standard diet. NEW & NOTEWORTHY Exercise training is beneficial in patients with heart failure (HF) or diabetes. However, less is known of the possible benefit of exercise training for HF patients with diabetes. Using a rat model where both diabetes and MI had been induced, we showed that 2 mo after MI, 8 wk of exercise training failed to improve cardiac function and metabolism in diabetic animals in contrast to lean animals.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Condicionamiento Físico Animal , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ecocardiografía , Metabolismo Energético , Corazón/fisiopatología , Masculino , Infarto del Miocardio/fisiopatología , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal , Estrés Fisiológico
14.
Circulation ; 111(23): 3025-33, 2005 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-15939817

RESUMEN

BACKGROUND: Life-threatening cardiac arrhythmia is a major source of mortality worldwide. Besides rare inherited monogenic diseases such as long-QT or Brugada syndromes, which reflect abnormalities in ion fluxes across cardiac ion channels as a final common pathway, arrhythmias are most frequently acquired and associated with heart disease. The mineralocorticoid hormone aldosterone is an important contributor to morbidity and mortality in heart failure, but its mechanisms of action are incompletely understood. METHODS AND RESULTS: To specifically assess the role of the mineralocorticoid receptor (MR) in the heart, in the absence of changes in aldosteronemia, we generated a transgenic mouse model with conditional cardiac-specific overexpression of the human MR. Mice exhibit a high rate of death prevented by spironolactone, an MR antagonist used in human therapy. Cardiac MR overexpression led to ion channel remodeling, resulting in prolonged ventricular repolarization at both the cellular and integrated levels and in severe ventricular arrhythmias. CONCLUSIONS: Our results indicate that cardiac MR triggers cardiac arrhythmias, suggesting novel opportunities for prevention of arrhythmia-related sudden death.


Asunto(s)
Arritmias Cardíacas/etiología , Regulación de la Expresión Génica/fisiología , Miocardio/metabolismo , Receptores de Mineralocorticoides/genética , Animales , Arritmias Cardíacas/patología , Calcio/metabolismo , Enfermedad Crítica , Muerte Súbita , Modelos Animales de Enfermedad , Electrocardiografía , Electrofisiología , Humanos , Canales Iónicos , Ratones , Ratones Transgénicos , Miocardio/patología , Miocitos Cardíacos/metabolismo , ARN Mensajero/análisis
15.
Artículo en Inglés | MEDLINE | ID: mdl-17083071

RESUMEN

BACKGROUND: Previous experiments have studied separately the development of either cardiac or aortic fibrosis and stiffness in aldosterone (Aldo)-salt hypertensive rats. Our aim was to determine in vivo the effects of Aldo and the Aldo receptor antagonist eplerenone (Epl) on simultaneous changes in cardiac and arterial structure and function and their interactions. METHODS AND RESULTS: Aldo was administered in uninephrectomised Sprague-Dawley rats receiving a high-salt diet from 8 to 12 weeks of age. Three groups of Aldo-salt rats were treated with 1 to 100 mg/kg-1. d-1 Epl by gavage. Arterial elasticity was measured by elastic modulus (Einc)-wall stress curves using medial cross-sectional area (MCSA). The cardiac and arterial walls were analysed by histomorphometry (elastin and collagen), immunohistochemistry (EIIIA fibronectin, Fn), and Northern blot (collagens I and III). Aldo caused increased systolic blood pressure (SBP), carotid Einc, MCSA, and EIIIA Fn with no change in wall stress or elastin and collagen densities. No difference in collagen mRNA levels was detected between groups. During the same period, cardiac mass and collagen mRNA and protein levels increased markedly in the myocardial tissue. Epl normalised collagen in the myocardium, Eincwall stress curves, MCSA, and EIIIA Fn in Aldo rats. These dose-dependent effects were not accompanied by a consistent reduction in SBP and cardiac mass. CONCLUSIONS: In exogenous hyperaldosteronism in the rat, Aldo causes independently myocardial collagen and arterial Fn accumulation, the latter being responsible for increased intrinsic carotid stiffness. Epl prevents both cardiac and arterial effects but does not reduce consistently SBP.


Asunto(s)
Cardiopatías/etiología , Cardiopatías/patología , Hipertensión/complicaciones , Hipertensión/patología , Espironolactona/análogos & derivados , Enfermedades Vasculares/etiología , Enfermedades Vasculares/patología , Animales , Aorta/patología , Presión Sanguínea/efectos de los fármacos , Northern Blotting , Enfermedades de las Arterias Carótidas/etiología , Enfermedades de las Arterias Carótidas/patología , Colágeno/metabolismo , Eplerenona , Fibrosis , Hipertensión/inducido químicamente , Inmunohistoquímica , Masculino , Antagonistas de Receptores de Mineralocorticoides , Miocardio/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Espironolactona/uso terapéutico , Relación Estructura-Actividad
16.
Hypertension ; 68(2): 392-400, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27296994

RESUMEN

Hypertension, which is a risk factor of heart failure, provokes adaptive changes at the vasculature and cardiac levels. Notch3 signaling plays an important role in resistance arteries by controlling the maturation of vascular smooth muscle cells. Notch3 deletion is protective in pulmonary hypertension while deleterious in arterial hypertension. Although this latter phenotype was attributed to renal and cardiac alterations, the underlying mechanisms remained unknown. To investigate the role of Notch3 signaling in the cardiac adaptation to hypertension, we used mice with either constitutive Notch3 or smooth muscle cell-specific conditional RBPJκ knockout. At baseline, both genotypes exhibited a cardiac arteriolar rarefaction associated with oxidative stress. In response to angiotensin II-induced hypertension, the heart of Notch3 knockout and SM-RBPJκ knockout mice did not adapt to pressure overload and developed heart failure, which could lead to an early and fatal acute decompensation of heart failure. This cardiac maladaptation was characterized by an absence of media hypertrophy of the media arteries, the transition of smooth muscle cells toward a synthetic phenotype, and an alteration of angiogenic pathways. A subset of mice exhibited an early fatal acute decompensated heart failure, in which the same alterations were observed, although in a more rapid timeframe. Altogether, these observations indicate that Notch3 plays a major role in coronary adaptation to pressure overload. These data also show that the hypertrophy of coronary arterial media on pressure overload is mandatory to initially maintain a normal cardiac function and is regulated by the Notch3/RBPJκ pathway.


Asunto(s)
Vasos Coronarios , Insuficiencia Cardíaca , Hipertensión/complicaciones , Músculo Liso Vascular , Receptor Notch3/metabolismo , Túnica Media , Adaptación Fisiológica , Animales , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Estrés Oxidativo , Transducción de Señal , Túnica Media/metabolismo , Túnica Media/patología
17.
Hypertension ; 67(3): 606-12, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26781273

RESUMEN

Galectin-3 (Gal-3) is involved in inflammation, fibrogenesis, and cardiac remodeling. Previous evidence shows that Gal-3 interacts with aldosterone in promoting macrophage infiltration and vascular fibrosis and that Gal-3 genetic and pharmacological inhibition prevents remodeling in a pressure-overload animal model of heart failure. We aimed to explore the contribution of Gal-3 and aldosterone in mechanisms leading to heart failure in a murine model. Male mice with cardiac-specific hyperaldosteronism underwent isoproterenol subcutaneous injections, to be then randomized to receive placebo, a Gal-3 inhibitor (modified citrus pectin [MCP]), an aldosterone antagonist (potassium canrenoate), or MCP+canrenoate for 14 days. Isoproterenol induced a rapid and persistent decrease in left ventricular fractional shortening (-20% at day 14); this was markedly improved by treatment with either MCP or canrenoate (both P<0.001 versus placebo). MCP and canrenoate also reduced cardiac hypertrophy and fibrosis and the expression of genes involved in fibrogenesis (Coll-1 and Coll-3) and macrophage infiltration (CD-68 and MCP-1). After isoproterenol, Gal-3 gene expression (P<0.05 versus placebo) and protein levels (-61% and -69% versus placebo) were decreased by both canrenoate and MCP. The combined use of antagonists of Gal-3 and aldosterone resulted in more pronounced effects on cardiac hypertrophy, inflammation, and fibrosis, when compared with either MCP or canrenoate alone. Inhibition of Gal-3 and aldosterone can reverse isoproterenol-induced left ventricular dysfunction, by reducing myocardial inflammation and fibrogenesis. Gal-3 likely participates in mechanisms of aldosterone-mediated myocardial damage in a heart failure murine model with cardiac hyperaldosteronism. Gal-3 inhibition may represent a new promising therapeutic option in heart failure.


Asunto(s)
Galectina 3/genética , Regulación de la Expresión Génica , Ventrículos Cardíacos/patología , ARN/genética , Disfunción Ventricular Izquierda/genética , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Fibrosis , Galectina 3/antagonistas & inhibidores , Galectina 3/biosíntesis , Ventrículos Cardíacos/fisiopatología , Isoproterenol/toxicidad , Masculino , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/diagnóstico
18.
Circulation ; 106(22): 2848-53, 2002 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-12451013

RESUMEN

BACKGROUND: Previous studies have demonstrated the development of cardiac fibrosis in aldosterone (Aldo)-salt hypertensive rats. Our aim was to determine the effects of Aldo and the Aldo receptor antagonist eplerenone (Epl) on in vivo mechanical properties of the carotid artery using echo-tracking system. METHODS AND RESULTS: Aldo was administered (1 microg/h) in uninephrectomized Sprague-Dawley rats (SD) receiving a high-salt diet from 8 to 12 weeks of age. Uninephrectomized control SD rats received a normal salt diet without Aldo. Three groups of Aldo-salt rats were treated with 1, 10, or 30 mg/kg(-1) x d(-1) of Epl by gavage. Elasticity was measured by elastic modulus (Einc)-wall stress curves using medial cross-sectional area (MCSA). The structure of the arterial wall was analyzed by histomorphometry (elastin and collagen), immunohistochemistry (EIIIA fibronectin, Fn), and Northern blot (collagens I and III). Aldo produced increased systolic arterial pressure, pulse pressure, Einc, MCSA, and EIIIA Fn with no change in wall stress or elastin and collagen densities compared with controls without Aldo. No differences in collagen mRNA levels were detected between groups. Epl blunted the increase in pulse pressure in Aldo rats and normalized Einc-wall stress curves, MCSA, and EIIIA Fn. These effects were dose dependent and not accompanied by a reduction in wall stress. CONCLUSIONS: Aldo is able to increase arterial stiffness associated with Fn accumulation, independently of wall stress. The preventive effects of Epl suggest a direct role for mineralocorticoid receptors in mechanical and structural alterations of large vessels in rat hyperaldosteronism.


Asunto(s)
Aldosterona/farmacología , Arterias Carótidas/metabolismo , Fibronectinas/metabolismo , Sodio en la Dieta/farmacología , Espironolactona/análogos & derivados , Espironolactona/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Northern Blotting , Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/efectos de los fármacos , Colágeno/biosíntesis , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Colágeno Tipo III/biosíntesis , Colágeno Tipo III/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Elasticidad/efectos de los fármacos , Elastina/biosíntesis , Eplerenona , Fibronectinas/análisis , Hiperaldosteronismo/inducido químicamente , Hiperaldosteronismo/tratamiento farmacológico , Hiperaldosteronismo/metabolismo , Inmunohistoquímica , Inyecciones Subcutáneas , Masculino , Antagonistas de Receptores de Mineralocorticoides , Nefrectomía , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Ultrasonografía , Grado de Desobstrucción Vascular/efectos de los fármacos
19.
Circulation ; 109(16): 1933-7, 2004 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15078792

RESUMEN

BACKGROUND: We analyzed the role of aldosterone in ischemia-induced neovascularization and the involvement of angiotensin II (Ang II) signaling in this effect. METHODS AND RESULTS: Ischemia was induced by right femoral artery ligature in mice treated or not with aldosterone (4.5 microg/day), aldosterone plus spironolactone (aldosterone receptor blocker; 20 mg/kg per day), or aldosterone plus valsartan (angiotensin type 1 [AT1] receptor blocker; 20 mg/kg per day). After 21 days, neovascularization was evaluated by microangiography, capillary density measurement, and laser-Doppler perfusion imaging. Protein level of vascular endothelial growth factor (VEGF) was determined by Western blot analysis in hindlimbs. mRNA levels of renin-angiotensin system components were also assessed by semiquantitative reverse transcription-polymerase chain reaction. Angiographic score, capillary number, and foot perfusion were improved in ischemic/nonischemic leg ratio by 1.4-, 1.5-, and 1.4-fold, respectively, in aldosterone-treated mice compared with controls (P<0.05). Aldosterone proangiogenic effect was associated with 2.3-fold increase in VEGF protein content (P<0.05). Treatments with spironolactone or with neutralizing VEGF antibody hampered the proangiogenic effect of aldosterone (P<0.05 versus aldosterone-treated mice). Interestingly, AT1 receptor blockade completely abrogated the aldosterone proangiogenic effect, emphasizing the involvement of Ang II-related pathway in aldosterone-induced vessel growth. In this view, angiotensinogen mRNA content was 2.2-fold increased in aldosterone-treated mice in reference to controls (P<0.05), whereas that of renin, angiotensin-converting enzyme, and AT1 receptor subtype was unaffected. Aldosterone treatment also decreased AT2 mRNA content by 2-fold (P<0.05 versus controls), suggesting that aldosterone may switch the Ang II pathway toward activation of vessel growth. CONCLUSIONS: This study shows for the first time that aldosterone increases neovascularization in the setting of ischemia through activation of Ang II signaling.


Asunto(s)
Aldosterona/farmacología , Inductores de la Angiogénesis/farmacología , Angiotensina II/metabolismo , Isquemia/tratamiento farmacológico , Neovascularización Fisiológica , Aldosterona/uso terapéutico , Inductores de la Angiogénesis/uso terapéutico , Animales , Vasos Sanguíneos/efectos de los fármacos , Isquemia/diagnóstico por imagen , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Radiografía , Flujo Sanguíneo Regional/efectos de los fármacos , Sistema Renina-Angiotensina , Transducción de Señal , Piel/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Circulation ; 110(13): 1819-25, 2004 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-15364804

RESUMEN

BACKGROUND: Elevated circulating aldosterone level is associated with impaired cardiovascular function. Although the mechanisms are not fully understood, aldosterone antagonists decrease total and cardiovascular mortality in heart failure and myocardial infarction. Aldosterone induces cardiac fibrosis in experimental models, and it is synthesized locally in rat heart. These observations suggest pathological effects of aldosterone in heart that remain unclear. METHODS AND RESULTS: Transgenic mice (TG) that overexpress the terminal enzyme of aldosterone biosynthesis, aldosterone synthase (AS), in heart have been raised by gene targeting with the alpha-myosin heavy chain promoter. AS mRNA increased 100-fold and aldosterone concentration 1.7-fold in hearts of male TG mice relative to wild-type. No structural or myocardial alterations were evidenced, because ventricle/body weight, AT1 and AT2 receptor binding, and collagen content were unchanged in TG. No alteration in cardiac function was evidenced by echocardiography, isolated perfused heart, or whole-cell patch clamp experiments. In contrast, coronary function was impaired, because basal coronary flow was decreased in isolated perfused heart (-55% of baseline values), and vasodilatation to acetylcholine, bradykinin, and sodium nitroprusside was decreased by 75%, 60%, and 75%, respectively, in TG mice compared with wild-type, showing that the defect was not related to NO production. CONCLUSIONS: Increased cardiac aldosterone production in male mice induces a major coronary endothelium-independent dysfunction with no detectable alterations in cardiac structure and function. However, coronary dysfunction may be harmful for coronary adaptation to increased flow demand.


Asunto(s)
Aldosterona/biosíntesis , Vasos Coronarios/patología , Citocromo P-450 CYP11B2/fisiología , Endotelio Vascular/patología , Miocardio/metabolismo , Acetilcolina/farmacología , Animales , Bradiquinina/farmacología , Calcio/metabolismo , Colágeno/biosíntesis , Circulación Coronaria , Vasos Coronarios/metabolismo , Citocromo P-450 CYP11B2/genética , Endotelio Vascular/metabolismo , Canales Iónicos/metabolismo , Transporte Iónico , Masculino , Ratones , Ratones Transgénicos , Óxido Nítrico/biosíntesis , Nitroprusiato/farmacología , Especificidad de Órganos , Técnicas de Placa-Clamp , Potasio/metabolismo , ARN Mensajero/biosíntesis , Ratas , Receptores de Angiotensina/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transgenes , Vasodilatación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA