Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Chin J Physiol ; 64(2): 72-79, 2021.
Article En | MEDLINE | ID: mdl-33938817

Long-term deprivation of female sex hormones has been shown to mediate accumulation of damaged mitochondria in ventricular muscle leading to cardiovascular dysfunction. Therefore, the roles of female sex hormones in mitochondrial quality control are closely focused. In the present study, depletion of female sex hormones impairing mitochondrial autophagy in the heart was hypothesized. Cardiac mitophagy was therefore investigated in the heart of 10-week ovariectomized (OVX) and sham-operated (SHAM) rats. By using isolated mitochondria preparation, results demonstrated an increase in mitochondrial PTEN-induced kinase 1 accumulation in the sample of OVX rats indicating mitochondrial outer membrane dysfunction. However, no change in p62 and LC3-II translocation to mitochondria was observed between two groups indicating unresponsiveness of mitophagosome formation in the OVX rat heart. This loss might be resulted from significant decreases in Parkin and Bcl2l13 expression, but not Bnip3 activation. In summary, results suggest that mitochondrial abnormality in the heart after deprivation of female sex hormones could consequently be due to desensitization of mitophagy process.


Mitochondria , Mitophagy , Animals , Autophagy , Female , Gonadal Steroid Hormones , Heart , Rats
2.
Life Sci ; 261: 118342, 2020 Nov 15.
Article En | MEDLINE | ID: mdl-32853655

AIMS: The increased incidence of heart failure with reduced ejection fraction in men compared with women suggests that male sex hormones significantly impact myocardial contractile activation. This study aims to examine associations among molecular alterations, cellular modulations and in vivo cardiac contractile function upon deprivation of testicular hormones. MAIN METHODS: Myocardial structure and functions were compared among sham-operated control and twelve-week orchidectomized (ORX) male rats with and without testosterone supplementation. KEY FINDINGS: Echocardiography and pressure-volume relationships demonstrated a decreased left ventricular ejection fraction compared with sham-operated controls. The percentage of contractility reduction was generally similar to the decrease in tension development detected in both right ventricular trabeculae and skinned isolated left ventricular cardiomyocytes of ORX rats. Reductions in tension cost and the rate constant of tension redevelopment (ktr) in ORX samples suggested a decrease in the rate of cross-bridge formation, reflecting a reduced number of cross-bridges. Slow cross-bridge detachment in ORX rat hearts could result from a shift of myosin heavy chain isoforms towards a slower ATPase activity ß-isoform and reductions in the phosphorylation levels of cardiac troponin I and myosin binding protein-C. All the changes in the ORX rat heart, including ejection fractions and myofilament protein expression and phosphorylation, were completed attenuated by a physiological dose of testosterone. SIGNIFICANCE: Testosterone plays a critical role in regulating the mechanical and contractile dynamics of the heart. Deprivation of male sex hormones cause the loss of normal preserved cardiac contractile function leading to a high risk of severe cardiomyopathy progression.


Cardiomyopathies/physiopathology , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Testosterone/metabolism , Animals , Disease Progression , Heart/physiology , Male , Myosin Heavy Chains/metabolism , Orchiectomy , Rats , Rats, Sprague-Dawley , Stroke Volume/physiology , Testosterone/administration & dosage , Testosterone/pharmacology , Ventricular Function, Left/physiology
3.
Free Radic Biol Med ; 160: 239-245, 2020 11 20.
Article En | MEDLINE | ID: mdl-32763410

A rise in heart disease incidence in women after menopause has led to investigations into the role of female sex hormones on cardiac function. Although various adverse changes in cardiac contractile function following loss of female sex hormones have been reported, a clear mechanism of action has never been characterized. In order to examine whether an elevation in oxidative stress is a major cause of cardiac contractile dysfunction after female sex hormone deprivation, cardiac functions of ovariectomized rats with and without supplementation of superoxide scavenger tempol were compared to those of sham-operated controls. Chronic deprivation of female sex hormones reduced total oxidative capacity and increased plasma carbonyl protein content. Tempol supplementation of ovariectomized rats significantly ameliorated plasma oxidative stress status. Echocardiography demonstrated a significant decrease in left ventricular ejection fraction in ovariectomized rats, which was completely prevented by tempol supplementation. Decreased myocardial contractility occurs with reduced maximum myofilament force of contraction and amplitude of transient intracellular Ca2+ concentration, both phenomena completely attenuated by tempol supplementation. However, tempol only partially prevented shift of heart myosin heavy chain from dominant α-to ß-isoform of ovariectomized rats. Immunoblot analysis of protein carbonylation indicated that tempol supplementation significantly reduced the level of cardiac myofibrillar proteins oxidation increased in ovariectomized rat heart. Taken together, the results indicate changes of cardiac contractile machinery following loss of female sex hormones were, in part, due to an increase in oxidative stress, and antioxidant supplementation could be considered another potential prevention measure in postmenopausal women.


Antioxidants , Ventricular Function, Left , Animals , Cyclic N-Oxides/pharmacology , Female , Oxidative Stress , Rats , Spin Labels , Stroke Volume
4.
BMC Complement Med Ther ; 20(1): 140, 2020 May 06.
Article En | MEDLINE | ID: mdl-32375750

BACKGROUND: Ecdysteroids are polyhydroxylated steroids present in invertebrates and plants. 20-Hydroxyecdysone (20E) is the most common and the main biologically active compound of ecdysteroids. Previous studies have demonstrated anabolic and metabolic effects of 20E in mammals. However, it is unknown whether 20E has a positive effect on all aspects of cardiometabolic syndrome. The aims of this study were to investigate the favorable effect and possible underlying mechanisms of 20E in a rat model of cardiometabolic syndrome (CMS) induced by a high-calorie diet combined with female sex hormone deprivation. METHODS: 20E (5 mg/kg, 10 mg/kg, or 20 mg/kg) or pioglitazone (PIO) (10 mg/kg) was intragastrically administered to sham-operated Sprague-Dawley female rats and ovariectomized rats fed a high-fat-high-fructose diet (OHFFD) for 8 weeks. The phenotypic characteristics of CMS, including central adiposity, blood pressure, serum lipid profile, glucose tolerance, insulin action on skeletal muscle glucose transport activity and hepatic protein expression, were determined. RESULTS: Some CMS characteristics were improved by 20E treatment. Rats treated with 20E had lower body weight, abdominal fat accumulation than rats treated with vehicle control without changes in total caloric intake and fat-free mass. OHFFD rats exhibited high blood pressure, but 20E-treated rats maintained normal blood pressure with a lower level of low-density lipoprotein (LDL)-cholesterol. Although 20E showed no positive effect on inducing insulin-mediated glucose transport in the skeletal muscle of OHFFD rats, 20E improved whole body glucose homeostasis. Analysis of protein expression in livers from 20E-treated rats revealed significantly increased expression of pAkt Ser473, pFOXO1 Ser256, pAMPKα Thr172, and FGF21. CONCLUSION: 20E treatment can alleviate cardiometabolic disorder caused by a high-fat-high-fructose diet and female sex hormone deprivation. In particular, 20E helps improve whole body insulin sensitivity in OHFFD rats, and the mechanisms that underlie this favorable effect are potentially mediated by the activation of AMPK and FGF21. The present study indicates that 20E could be an alternative therapeutic option for the prevention and alleviation of cardiometabolic syndrome.


Blood Pressure/drug effects , Ecdysterone/pharmacology , Metabolic Syndrome/drug therapy , Ovariectomy , Animals , Diet, High-Fat , Disease Models, Animal , Female , Fructose/administration & dosage , Rats , Rats, Sprague-Dawley
5.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R829-R842, 2020 05 01.
Article En | MEDLINE | ID: mdl-32159365

Cardiac inflammation has been proposed as one of the primary mechanisms of anthracycline-induced acute cardiotoxicity. A reduction in cardiac inflammation might also reduce cardiotoxicity. This study aimed to evaluate the potential of estrogen therapy and regular exercise on attenuating cardiac inflammation in the context of doxorubicin-induced cardiomyopathy. Ovariectomized rats were randomly allocated into estrogen supplementation, exercise training, and mast cell stabilizer treatment groups. Eight weeks after ovariectomy, rats received six cumulative doses of doxorubicin for two weeks. Echocardiography demonstrated a progressive decrease in ejection fraction in doxorubicin-treated rats without hypertrophic effect. This systolic defect was completely prevented by either estrogen supplementation or mast cell stabilizer treatment but not by regular exercise. As a heart disease indicator, increased ß-myosin heavy chain expression induced by doxorubicin could only be prevented by estrogen supplementation. Decrease in shortening and intracellular Ca2+ transients of cardiomyocytes were due to absence of female sex hormones without further effects of doxorubicin. Again, estrogen supplementation and mast cell stabilizer treatment prevented these changes but exercise training did not. Histological analysis indicated that the hyperactivation of cardiac mast cells in ovariectomized rats was augmented by doxorubicin. Estrogen supplementation and mast cell stabilizer treatment completely prevented both increases in mast cell density and degranulation, whereas exercise training partially attenuated the hyperactivation. Our results, therefore, suggest that estrogen supplementation acts similarly to mast cell stabilizers in attenuating the effects of doxorubicin. Ineffectiveness of regular exercise in preventing the acute cardiotoxicity of doxorubicin might be due to a lesser effect on preventing cardiac inflammation.


Cell Degranulation/drug effects , Doxorubicin , Estradiol/administration & dosage , Estrogen Replacement Therapy , Exercise Therapy , Mast Cells/drug effects , Myocardial Contraction/drug effects , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Animals , Calcium Signaling/drug effects , Cardiotoxicity , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Mast Cells/metabolism , Mast Cells/pathology , Myocardium/metabolism , Myocardium/pathology , Ovariectomy , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
6.
Physiol Rep ; 7(14): e14192, 2019 07.
Article En | MEDLINE | ID: mdl-31353833

Androgen therapy provides cardiovascular benefits for hypogonadism. However, myocardial hypertrophy, fibrosis, and infarction have been reported in testosterone or androgenic anabolic steroid abuse. Therefore, better understanding of the factors leading to adverse results of androgen abuse is needed. The aim of the present study was to examine the impact of high dose of androgen treatment on cardiac biology, and whether exposure duration modulates this response. Male rats were treated with 10 mg/kg testosterone, three times a week, for either 4 or 12 weeks; vehicle injections served as controls. Four weeks of testosterone treatment induced an increase in ventricular wall thickness, indicative of concentric hypertrophy, as well as increased ejection fraction; in contrast, both parameters were blunted following 12 weeks of high-dose testosterone treatment. Cardiac myocyte contractile parameters were assessed in isolated electrically stimulated myocytes (sarcomere and intracellular calcium dynamics), and in chemically permeabilized isolated myocardium (myofilament force development and tension-cost). High-dose testosterone treatment for 4 weeks was associated with increased myocyte contractile parameters, while 12 weeks treatment induced significant depression of these parameters, mirroring the cardiac pump function results. In conclusion, chronic administration of high-dose testosterone initially induces increased cardiac function. However, this initial beneficial impact is followed by significant depression of cardiac pump function, myocyte contractility, and cardiac myofilament function. Our results indicate that chronic high-testosterone usage is of limited use and may, instead, induce significant cardiac dysfunction.


Androgens/pharmacology , Heart/drug effects , Myocardial Contraction , Testosterone/pharmacology , Androgens/administration & dosage , Androgens/adverse effects , Animals , Calcium/metabolism , Cells, Cultured , Heart/physiology , Male , Rats , Rats, Sprague-Dawley , Sarcomeres/drug effects , Sarcomeres/metabolism , Sarcomeres/physiology , Testosterone/administration & dosage , Testosterone/adverse effects
7.
Life Sci ; 219: 209-218, 2019 Feb 15.
Article En | MEDLINE | ID: mdl-30658099

AIMS: Regular exercise is recommended in postmenopausal women to prevent the development of heart disease, but mechanism underlying the protection is not completely understood. Many studies have suggested that exercise training notably mediated whole body immune and inflammatory functions. Whether exercise training prevents cardiac dysfunction after deprivation of female sex hormones by inhibiting cardiac immune activation is therefore interesting. MAIN METHODS: Nine-week treadmill running program was introduced in sham-operated and ovariectomized rats. In addition, chronic angiotensin II infusion was further challenged to activate pathological cardiac remodeling. Cardiac remodeling in associated with the density and degranulation of cardiac mast cells was then evaluated. KEY FINDINGS: With exogenous angiotensin II-induced hypertension, cardiac hypertrophy with myocardial fibrosis was shown similarly in both sham-operated controls and ovariectomized rats. Although exercise training did not prevent cardiac hypertrophy, myocardial fibrosis was abolished by exercise. While ovariectomy increased both cardiac mast cell density and degranulation percentage, angiotensin II infusion only enhanced mast cell density. Exercise training could not decrease the density of mast cells, but it did normalize the percentage of degranulation in all groups. Correlation analysis suggested that cardiac mast cell activation is inversely associated with cardiomyocyte hypertrophy due to exercise training but is directly correlated to cardiac hypertrophy by angiotensin II infusion. SIGNIFICANCE: Exercise training could attenuate cardiac mast cell hyperactivation induced by either deprivation of female sex hormones or excessive angiotensin II. Additionally, cardiac mast cells could be a solution in the distinction between physiological and pathological hypertrophic development.


Angiotensin II/pharmacology , Mast Cells/physiology , Physical Conditioning, Animal/physiology , Ventricular Remodeling/physiology , Animals , Female , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Ovariectomy , Rats , Rats, Sprague-Dawley , Ventricular Remodeling/drug effects
8.
Am J Physiol Heart Circ Physiol ; 316(2): H360-H370, 2019 02 01.
Article En | MEDLINE | ID: mdl-30499711

Here, we aimed to explore sex differences and the impact of sex hormones on cardiac contractile properties in doxorubicin (DOX)-induced cardiotoxicity. Male and female Sprague-Dawley rats were subjected to sham surgery or gonadectomy and then treated or untreated with DOX (2 mg/kg) every other week for 10 wk. Estrogen preserved maximum active tension (Tmax) with DOX exposure, whereas progesterone and testosterone did not. The effects of sex hormones and DOX correlated with both altered myosin heavy chain isoform expression and myofilament protein oxidation, suggesting both as possible mechanisms. However, acute treatment with oxidative stress (H2O2) or a reducing agent (DTT) indicated that the effects on Tmax were mediated by reversible myofilament oxidative modifications and not only changes in myosin heavy chain isoforms. There were also sex differences in the DOX impact on myofilament Ca2+ sensitivity. DOX increased Ca2+ sensitivity in male rats only in the absence of testosterone and in female rats only in the presence of estrogen. Conversely, DOX decreased Ca2+ sensitivity in female rats in the absence of estrogen. In most instances, this mechanism was through altered phosphorylation of troponin I at Ser23/Ser24. However, there was an additional DOX-induced, estrogen-dependent, irreversible (by DTT) mechanism that altered Ca2+ sensitivity. Our data demonstrate sex differences in cardiac contractile responses to chronic DOX treatment. We conclude that estrogen protects against chronic DOX treatment in the heart, preserving myofilament function. NEW & NOTEWORTHY We identified sex differences in cardiotoxic effects of chronic doxorubicin (DOX) exposure on myofilament function. Estrogen, but not testosterone, decreases DOX-induced oxidative modifications on myofilaments to preserve maximum active tension. In rats, DOX exposure increased Ca2+ sensitivity in the presence of estrogen but decreased Ca2+ sensitivity in the absence of estrogen. In male rats, the DOX-induced shift in Ca2+ sensitivity involved troponin I phosphorylation; in female rats, this was through an estrogen-dependent mechanism.


Antioxidants/pharmacology , Doxorubicin/toxicity , Estrogens/pharmacology , Papillary Muscles/metabolism , Testosterone/pharmacology , Animals , Calcium/metabolism , Cardiotoxicity , Estrogens/metabolism , Female , Male , Myocardial Contraction , Myofibrils/drug effects , Myofibrils/metabolism , Myofibrils/physiology , Oxidative Stress , Papillary Muscles/drug effects , Papillary Muscles/physiology , Phosphorylation , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Sex Factors , Testosterone/metabolism , Troponin I/metabolism
9.
Steroids ; 126: 79-84, 2017 10.
Article En | MEDLINE | ID: mdl-28803209

BACKGROUND: Ecdysteroids, a group of steroid hormones found in insects and many plants, have been shown to prevent various changes in mammalian tissues after female sex hormone deprivation. PURPOSE: To examine whether an ecdysteroid, 20-hydroxyecdysone (20-HE), exhibits regulatory or protective roles in the cardiovascular system. STUDY DESIGN/METHOD: Blood pressure and cardiac function were evaluated in spontaneously hypertensive rats (SHR) during and after daily treatment with 20-HE for six weeks. RESULTS: The progressive increase in systolic blood pressure with age in SHR rats was significantly lower in animals treated with either 5 or 10mg/kg body weight of 20-HE. However, treatment with 20-HE did not diminish the increase in diastolic pressure. Echocardiography after six weeks of treatment demonstrated that the left ventricular chamber of SHR rats treated with 20-HE was smaller than that of SHR controls, while contractility was not affected by 20-HE. Histological images also demonstrated a decrease in cardiomyocyte cross-sectional area in 20-HE treated groups. Interestingly, treatment with 20-HE caused a shift in cardiac myosin heavy chain towards more ß-isoforms. SHR rats treated with 20-HE also exhibited a decrease in seminal vesicular weight and an increase in testicular weight, especially at a dose of 10mg/kg body weight. This finding suggests a possible anti-androgenic effect of 20-HE. CONCLUSION: Our finding reveal that 20-HE has a beneficial effect on reducing blood pressure and consequently preventing dilated cardiac hypertrophy in SHR rats.


Ecdysterone/pharmacology , Heart/drug effects , Heart/physiology , Myocardium/pathology , Animals , Blood Pressure/drug effects , Heart/physiopathology , Male , Rats , Rats, Inbred SHR
10.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Article En | MEDLINE | ID: mdl-28590578

The benefits of α-mangostin for various tissues have been reported, but its effect on the heart has not been clarified. This study aimed to evaluate the effects of α-mangostin on cardiac function. Using a cardiac sarcoplasmic reticulum (SR) membrane preparation, α-mangostin inhibited SR Ca2+ -ATPase activity in a dose-dependent manner (IC50 of 6.47 ± 0.7 µM). Its suppressive effect was specific to SR Ca2+ -ATPase but not to myofibrillar Ca2+ -ATPase. Using isolated cardiomyocytes, 50 µM of α-mangostin significantly increased the duration of cell relengthening and increased the duration of Ca2+ transient decay, suggesting altered myocyte relaxation. The relaxation effect of α-mangostin was also supported in vivo after intravenous infusion. A significant suppression of both peak pressure and rate of ventricular relaxation (-dP/dt) relative to DMSO infusion was observed. The results from the present study demonstrated that α-mangostin exerts specific inhibitory action on SR Ca2+ -ATPase activity, leading to myocardial relaxation dysfunction.


Diastole/drug effects , Heart Ventricles/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Xanthones/toxicity , Animals , Heart Ventricles/physiopathology , Male , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
11.
J Endocrinol ; 232(3): 547-560, 2017 03.
Article En | MEDLINE | ID: mdl-28096436

Angiotensin II (ANGII) is reportedly involved in the development of skeletal muscle insulin resistance. The present investigation evaluated the effects of two ANGII doses on the phenotypic characteristics of insulin resistance syndrome and insulin action and signaling in rat skeletal muscle. Male Sprague-Dawley rats were infused with either saline (SHAM) or ANGII at a commonly used pressor dose (100 ng/kg/min; ANGII-100) or a higher pressor dose (500 ng/kg/min; ANGII-500) via osmotic minipumps for 14 days. We demonstrated that ANGII-100-infused rats exhibited the phenotypic features of non-obese insulin resistance syndrome, including hypertension, impaired glucose tolerance and insulin resistance of glucose uptake in the soleus muscle, whereas ANGII-500-treated rats exhibited diabetes-like symptoms, such as post-prandial hyperglycemia, impaired insulin secretion and hypertriglyceridemia. At the cellular level, insulin-stimulated glucose uptake in the soleus muscle of the ANGII-100 group was 33% lower (P < 0.05) than that in the SHAM group and was associated with increased insulin-stimulated IRS-1 Ser307 and decreased Akt Ser473 and AS160 Thr642 phosphorylation and GLUT-4 expression. However, ANGII-500 infusion did not induce skeletal muscle insulin resistance or impair insulin signaling elements as initially anticipated. Moreover, we found that insulin-stimulated glucose uptake in the ANGII-500 group was accompanied by the enhanced expression of ACE2 and MasR proteins, which are the key elements in the non-classical pathway of the renin-angiotensin system. Collectively, this study demonstrates for the first time that chronic infusion with these two pressor doses of ANGII induced differential metabolic responses at both the systemic and skeletal muscle levels.


Angiotensin II/pharmacology , Insulin/metabolism , Muscle, Skeletal/drug effects , Signal Transduction/drug effects , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Eating/drug effects , Glucose Transporter Type 4/metabolism , Insulin Resistance/physiology , Male , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
12.
J Endocrinol ; 229(1): 13-23, 2016 04.
Article En | MEDLINE | ID: mdl-26850730

Testosterone and androgenic anabolic steroids have been misused for enhancement of physical performance despite many reports on cardiac sudden death. Although physiological level of testosterone provided many regulatory benefits to human health, including the cardiovascular function, supra-physiological levels of the hormone induce hypertrophy of the heart with unclear contractile activation. In this study, dose- and time-dependent effects of high-testosterone treatment on cardiac structure and function were evaluated. Adult male rats were divided into four groups of testosterone treatment for 0, 5, 10, and 20 mg/kg BW for 4, 8, or 12 weeks. Increases in both percentage heart:body weight ratio and cardiomyocyte cross-sectional area in representing hypertrophy of the heart were significantly shown in all testosterone-treated groups to the same degree. In 4-week-treated rats, physiological cardiac hypertrophy was apparent with an upregulation of α-MHC without any change in myofilament contractile activation. In contrast, pathological cardiac hypertrophy was observed in 8- and 12-week testosterone-treated groups, as indicated by suppression of myofilament activation and myocardial collagen deposition without transition of MHC isoforms. Only in 12-week testosterone-treated group, eccentric cardiac hypertrophy was demonstrated with unaltered myocardial stiffness, but significant reductions in the phosphorylation signals of ERK1/2 and mTOR. Results of our study suggest that the outcome of testosterone-induced cardiac hypertrophy is not dose dependent but is rather relied on the factor of exposure to duration in inducing maladaptive responses of the heart.


Cardiomegaly/chemically induced , Heart/drug effects , Testosterone/administration & dosage , Testosterone/adverse effects , Animals , Male , Random Allocation , Rats, Sprague-Dawley
13.
J Physiol Sci ; 66(2): 165-73, 2016 Mar.
Article En | MEDLINE | ID: mdl-26467449

It is well accepted that regular exercise is a significant factor in the prevention of cardiac dysfunction; however, the cardioprotective mechanism is as yet not well defined. We have examined whether regular exercise can modulate the activity of cardiac mast cells (CMC) after deprivation of female sex hormones, as well as the density and percentage degranulation of mast cells, in ventricular tissue of ovariectomized (OVX) rats after an 11-week running program. A significant increase in CMC density with a greater percentage degranulation was induced after ovarian sex hormone deprivation. Increased CMC density was prevented by estrogen supplements, but not by regular training. To the contrary, increased CMC degranulation in the OVX rat heart was attenuated by exercise training, but not by estrogen supplement. These findings indicate a significant correlation between the degree of CMC degranulation and myocyte cross-section area. However, no change in the expression of inflammatory mediators, including chymase, interleukin-6, and interleukin-10, was detected. Taken together, these results clearly indicate one of the cardioprotective mechanisms of regular aerobic exercise is the modulation of CMC activation.


Heart Ventricles/physiopathology , Mast Cells/physiology , Physical Conditioning, Animal/physiology , Animals , Chymases/metabolism , Estrogens/metabolism , Female , Heart Ventricles/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Mast Cells/metabolism , Muscle Cells/metabolism , Muscle Cells/physiology , Myocardium/metabolism , Ovariectomy/methods , Rats , Rats, Sprague-Dawley
14.
J Steroid Biochem Mol Biol ; 147: 1-9, 2015 Mar.
Article En | MEDLINE | ID: mdl-25448746

Increased susceptibility to stress-induced myocardial damage is a significant concern in addition to decreased cardiac performance in postmenopausal females. To determine the potential mechanisms underlying myocardial vulnerability after deprivation of female sex hormones, cardiac mitochondrial function is determined in 10-week ovariectomized rats (OVX). Significant mitochondrial swelling in the heart of OVX rats is observed. This structural alteration can be prevented with either estrogen or progesterone supplementation. Using an isolated mitochondrial preparation, a decrease in ATP synthesis by complex I activation in an OVX rat is completely restored by estrogen, but not progesterone. At basal activation, reactive oxygen species (ROS) production from the mitochondria is not affected by the ovariectomy. However, after incubated in the presence of either high Ca(2+) or antimycin-A, there is a significantly higher mitochondrial ROS production in the OVX sample compared to the control. This increased stress-induced ROS production is not observed in the preparation isolated from the hearts of OVX rats with estrogen or progesterone supplementation. However, deprivation of female sex hormones has no effect on the protein expression of electron transport chain complexes, mitofusin 2, or superoxide dismutase 2. Taken together, these findings suggest that female sex hormones, estrogen and progesterone, play significant regulatory roles in maintaining normal mitochondrial properties by stabilizing the structural assembly of mitochondria as well as attenuating mitochondrial ROS production. Estrogen, but not progesterone, also plays an important role in modulating mitochondrial ATP synthesis.


Estrogens/metabolism , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Adenosine Triphosphate/biosynthesis , Animals , Estrogens/administration & dosage , Estrogens/pharmacology , Female , GTP Phosphohydrolases , Membrane Proteins/metabolism , Mitochondria, Heart/drug effects , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Ovariectomy , Progesterone/administration & dosage , Progesterone/metabolism , Progesterone/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
15.
J Physiol Sci ; 64(4): 269-77, 2014 Jul.
Article En | MEDLINE | ID: mdl-24777837

Ovariectomy leads to suppression of cardiac myofilament activation in healthy rats implicating the physiological essence of female sex hormones on myocardial contraction. However, the possible function of these hormones during pathologically induced myofilament adaptation is not known. In this study, sham-operated and ovariectomized female rats were chronically exposed to angiotensin II (AII), which has been shown to cause myocardial adaptation. In the shams, AII induced cardiac adaptation by increasing myofilament Ca(2+) sensitivity. Interestingly, this hypersensitivity was further enhanced in AII-infused ovariectomized rats. Ovariectomy increased the phosphorylation levels of cardiac tropomyosin, which may underlie the mechanism of hypersensitivity. On the other hand, AII infusion did not alter maximal tension that was suppressed after ovariectomy. This finding coincided with a comparable increase in ß-isoform of myosin heavy chains in both ovariectomized groups. Together, it is conceivable that female sex hormones serve as predominant factors that regulate cardiac myofilament activation. Furthermore, they may prevent stress-induced myofilament maladaptation.


Angiotensin II/pharmacology , Gonadal Steroid Hormones/metabolism , Heart/physiology , Myofibrils/metabolism , Myofibrils/physiology , Animals , Calcium/metabolism , Female , Heart/drug effects , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myosin Heavy Chains/metabolism , Ovariectomy/adverse effects , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Tropomyosin/metabolism
16.
Am J Physiol Heart Circ Physiol ; 304(3): H465-72, 2013 Feb 01.
Article En | MEDLINE | ID: mdl-23203968

Alterations in intracellular Ca(2+) transients of cardiomyocytes in orchidectomized (ORX) rats could be a cause of cardiac dysfunction in the hypogonadal condition. To investigate the role of male sex hormones in intracellular Ca(2+) homeostasis during relaxation, Ca(2+)-handling activities by sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) and the Na(+)/Ca(2+) exchanger (NCX) were evaluated in the ventricular muscle of 10-wk-old ORX rats with and without testosterone supplementation (2.5 mg/kg testosterone propionate, 2 times/wk). ORX induced a 50% decrease in contraction force accompanied by a prolonged time to achieve 50% relaxation (T(50)) in isolated intact ventricular trabeculae, which was partially corrected by testosterone administration. Maximum active tension was also suppressed in ORX rats without changes in myofilament Ca(2+) sensitivity and passive stiffness of the heart. Using a sarcoplasmic reticulum-enriched membrane preparation, the maximum thapsigargin-sensitive SERCA activity of the ORX rat was 27% lower with an increased Ca(2+) sensitivity, which was prevented by testosterone treatment. However, neither changes in SERCA content nor its modulating components, sarcolipin and heat shock protein 20, were detected in the ORX rat, but there was a significant decrease in the phosphorylated Thr(17) form of phospholamban. Despite a lower level of NCX protein in the heart of ORX rats, prolonged T(50) disappeared after an incubation with thapsigargin (10 µM), implying a lack of effect of male sex hormone deficiency on NCX function. These findings indicate that male sex hormones can regulate cardiac relaxation by acting mainly through SERCA. However, a detailed mechanism of SERCA modulation under male sex hormone deficiency status remains to be explored.


Myocardial Contraction/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Sodium-Calcium Exchanger/physiology , Testosterone/pharmacology , Animals , Blotting, Western , Body Weight/drug effects , Heart/drug effects , Male , Muscle Contraction/physiology , Muscle Proteins/pharmacology , Muscle, Skeletal/drug effects , Orchiectomy , Organ Size/drug effects , Proteolipids/pharmacology , Rats , Rats, Sprague-Dawley , Sarcomeres/drug effects , Sarcomeres/ultrastructure , Seminal Vesicles/drug effects , Testosterone/blood , Testosterone/physiology , Trabecular Meshwork/physiology
17.
PLoS One ; 7(8): e43179, 2012.
Article En | MEDLINE | ID: mdl-22912821

Endothelin (ET-1) is a peptide hormone mediating a wide variety of biological processes and is associated with development of cardiac dysfunction. Generally, ET-1 is regarded as a molecular marker released only in correlation with the observation of a hypertrophic response or in conjunction with other hypertrophic stress. Although the cardiac hypertrophic effect of ET-1 is demonstrated, inotropic properties of cardiac muscle during chronic ET-1-induced hypertrophy remain largely unclear. Through the use of a novel in vitro multicellular culture system, changes in contractile force and kinetics of rabbit cardiac trabeculae in response to 1 nM ET-1 for 24 hours can be observed. Compared to the initial force at t = 0 hours, ET-1 treated muscles showed a ~2.5 fold increase in developed force after 24 hours without any effect on time to peak contraction or time to 90% relaxation. ET-1 increased muscle diameter by 12.5 ± 3.2% from the initial size, due to increased cell width compared to non-ET-1 treated muscles. Using specific signaling antagonists, inhibition of NCX, CaMKII, MAPKK, and IP3 could attenuate the effect of ET-1 on increased developed force. However, among these inhibitions only IP3 receptor blocker could not prevent the increase muscle size by ET-1. Interestingly, though calcineurin-NFAT inhibition could not suppress the effect of ET-1 on force development, it did prevent muscle hypertrophy. These findings suggest that ET-1 provokes both inotropic and hypertrophic activations on myocardium in which both activations share the same signaling pathway through MAPK and CaMKII in associated with NCX activity.


Cardiomegaly/etiology , Endothelins/pharmacology , Myocardial Contraction/drug effects , Signal Transduction/drug effects , Analysis of Variance , Animals , Benzylamines , Boron Compounds , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclosporine , Immunoblotting , In Vitro Techniques , Indoles , Kinetics , Maleimides , Mitogen-Activated Protein Kinases/metabolism , NFATC Transcription Factors/metabolism , Rabbits , Sulfonamides , Thiourea/analogs & derivatives , Time Factors
18.
Am J Physiol Heart Circ Physiol ; 302(12): H2509-17, 2012 Jun 15.
Article En | MEDLINE | ID: mdl-22467304

Numerous studies have aimed to elucidate markers for the onset of decompensatory hypertrophy and heart failure in vivo and in vitro. Alterations in the force-frequency relationship are commonly used as markers for heart failure with a negative staircase being a hallmark of decompensated cardiac function. Here we aim to determine the functional and molecular alterations in the very early stages of compensatory hypertrophy through analysis of the force-frequency relationship, using a novel isolated muscle culture system that allows assessment of force-frequency relationship during the development of hypertrophy. New Zealand white male rabbit trabeculae excised from the right ventricular free wall were utilized for all experiments. Briefly, muscles held at constant preload and contracting isometrically were stimulated to contract in culture for 24 h, and in a subset up to 48 h. We found that, upon an increase in the preload and maintaining the muscles in culture for up to 24 h, there was an increase in baseline force produced by isolated trabeculae over time. This suggests a gradual compensatory response to the impact of increased preload. Temporal analysis of the force-frequency response during this progression revealed a significant blunting (at 12 h) and then reversal of the positive staircase as culture time increased (at 24 h). Phosphorylation analysis revealed a significant decrease in desmin and troponin (Tn)I phosphorylation from 12 to 24 h in culture. These results show that even very early on in the compensatory hypertrophy state, the force-frequency relationship is already affected. This effect on force-frequency relationship may, in addition to protein expression changes, be partially attributed to the alterations in myofilament protein phosphorylation.


Cardiomegaly/physiopathology , Heart/physiopathology , Myocardial Contraction/physiology , Myocardium/metabolism , Animals , Cardiomegaly/metabolism , Desmin/metabolism , Kinetics , Male , Phosphorylation , Rabbits , Tissue Culture Techniques , Troponin I/metabolism
19.
Am J Physiol Heart Circ Physiol ; 300(5): H1661-8, 2011 May.
Article En | MEDLINE | ID: mdl-21335468

A decrease in peak early diastolic filling velocity in postmenopausal women implies a sex hormone-related diastolic dysfunction. The regulatory effect of female sex hormones on cardiac distensibility therefore was evaluated in ovariectomized rats by determining the sarcomere length-passive tension relationship of ventricular skinned fiber preparations. Diabetes also was induced in the rat to assess the protective significance of female sex hormones on diastolic function. While ovariectomy had no effect on myocardial stiffness, collagen content, or titin ratio, a significant increase in myocardial stiffness was observed in diabetic rat only when female sex hormones were intact. The increased stiffness in diabetic-sham rats was accompanied by an elevated collagen content resulting from increases in the levels of procollagen and Smad2. Surprisingly, the increased myocardial stiffness in diabetic-sham rats was accompanied by a shift toward a more compliant N2BA of cardiac titin isoforms. The pCa-active tension relationship was analyzed at fixed sarcomere lengths of 2.0 and 2.3 µm to determine the magnitude of changes in myofilament Ca(2+) sensitivity between the two sarcomere lengths. Interestingly, high expression of N2BA titin was associated with a suppressed magnitude of changes in myofilament Ca(2+) sensitivity only in the diabetic-ovariectomized condition. Estrogen supplementation in diabetic-ovariectomized rats partially increased myocardial stiffness but completely reversed the change in myofilament Ca(2+) sensitivity. These results indicate a restrictive adaptation of myocardium governed by female sex hormones to maintain myofilament activity in compensation to the pathophysiological induction of cardiac dilatation by the diabetic condition.


Calcium Signaling/drug effects , Diabetes Mellitus, Experimental/physiopathology , Elasticity/drug effects , Estrogens/pharmacology , Heart/physiopathology , Myocardial Contraction/drug effects , Animals , Calcium Signaling/physiology , Collagen/metabolism , Connectin , Diabetes Mellitus, Experimental/metabolism , Diastole/physiology , Disease Models, Animal , Elasticity/physiology , Female , Muscle Proteins/metabolism , Myocardial Contraction/physiology , Ovariectomy , Procollagen/metabolism , Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Smad2 Protein/metabolism , Streptozocin
20.
J Appl Physiol (1985) ; 107(4): 1105-12, 2009 Oct.
Article En | MEDLINE | ID: mdl-19679745

The impact of regular exercise in protecting cardiac deteriorating results of female sex hormone deprivation was evaluated by measuring changes in intracellular Ca2+ removal activity of sarcoplasmic reticulum (SR) in ovariectomized rats following 9-wk treadmill running exercise at moderate intensity. Despite induction of cardiac hypertrophy in exercised groups of both sham-operated and ovariectomized rats, exercise training had no effect on SR Ca2+ uptake and SR Ca(2+)-ATPase (SERCA) in hormone intact rat heart. However, exercise training normalized the suppressed maximum SR Ca2+ uptake and SERCA activity in ovariectomized rat heart. While exercise training normalized the leftward shift in pCa (-log[Ca2+])-SR Ca2+ uptake relation in ovariectomized rats, no effect was detected in exercised sham-operated rats. Similar phenomena were also observed on SERCA and on phospholamban (PLB) phosphorylation levels; exercise training in ovariectomized rats enhanced SERCA expression to reach the level as that in sham-operated rats, in which there were no differences in SERCA and phospho-PLB levels between sedentary and exercised groups. In addition, the reduction in phospho-Thr(17) PLB in myocardium of ovariectomized rats was abolished by exercise training. These results showed that regular exercise maintains the molecular activation of cardiac SR Ca2+ uptake under normal physiological conditions and is able to induce a protective impact on cardiac SR Ca2+ uptake in ovarian sex hormone-deprived status.


Calcium/metabolism , Myocardium/enzymology , Ovariectomy , Physical Exertion , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/enzymology , Animals , Biological Transport , Calcium-Binding Proteins/metabolism , Calsequestrin , Carrier Proteins/metabolism , Female , Heart Ventricles/enzymology , Phosphorylation , Rats , Rats, Sprague-Dawley , Serine , Threonine
...