Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Chin J Physiol ; 64(2): 72-79, 2021.
Article in English | MEDLINE | ID: mdl-33938817

ABSTRACT

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.


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

ABSTRACT

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.


Subject(s)
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 in English | MEDLINE | ID: mdl-32763410

ABSTRACT

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.


Subject(s)
Antioxidants , Ventricular Function, Left , Animals , Cyclic N-Oxides/pharmacology , Female , Oxidative Stress , Rats , Spin Labels , Stroke Volume
4.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R829-R842, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32159365

ABSTRACT

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.


Subject(s)
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
5.
Physiol Rep ; 7(14): e14192, 2019 07.
Article in English | MEDLINE | ID: mdl-31353833

ABSTRACT

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.


Subject(s)
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
6.
Am J Physiol Heart Circ Physiol ; 316(2): H360-H370, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30499711

ABSTRACT

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.


Subject(s)
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
7.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28590578

ABSTRACT

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.


Subject(s)
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
8.
J Endocrinol ; 229(1): 13-23, 2016 04.
Article in English | MEDLINE | ID: mdl-26850730

ABSTRACT

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.


Subject(s)
Cardiomegaly/chemically induced , Heart/drug effects , Testosterone/administration & dosage , Testosterone/adverse effects , Animals , Male , Random Allocation , Rats, Sprague-Dawley
9.
J Physiol Sci ; 66(2): 165-73, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26467449

ABSTRACT

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.


Subject(s)
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
10.
J Appl Physiol (1985) ; 119(7): 831-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26272317

ABSTRACT

Data from the trial known as Testosterone in Older Men with Mobility Limitations (TOM) has indicated an association between testosterone administration and a greater risk for adverse cardiovascular events. We therefore propose that regular exercise is a cardioprotective alternative that prevents detrimental changes in contractile activation when a deficiency in male sex hormones exists. Ten-week-old orchidectomized (ORX) rats were subjected to a 9-wk treadmill running program at moderate intensity starting 1 wk after surgery. Although exercise-induced cardiac hypertrophy was observed both in rats that underwent ORX and sham surgery, regular exercise enhanced cardiac myofilament Ca(2+) sensitivity and myosin light-chain 2 phosphorylation only in rats that underwent a sham operation. Although the rats that had sham surgery and and given exercise exhibited no change in maximum developed tension, regular running prevented the suppression of maximum active tension in the hearts of ORX rats. Regular exercise also prevented a shift in myosin heavy chain (MHC) isoforms toward ß-MHC, a reduction in sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA) activity, and an increase in SERCA sensitivity in the hearts of ORX rats. Neither SERCA content nor its modulating component, phospholamban (PLB), was altered by exercise in either sham-operated or ORX rats. However, decreases in the phosphorylated Thr(17) form of PLB and the phosphorylated Thr(287) form of Ca(2+)/calmodulin-dependent kinase II in the hearts of ORX rats were abolished after regular exercise. These results thus support the use of regular running as a cardioprotective alternative to testosterone replacement in hypogonadal conditions.


Subject(s)
Myocardial Contraction/physiology , Myosin Heavy Chains/metabolism , Orchiectomy , Physical Conditioning, Animal/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiac Myosins/metabolism , Male , Myofibrils/metabolism , Myosin Light Chains/metabolism , Organ Size , Phosphorylation , Rats , Rats, Sprague-Dawley , Running
11.
J Steroid Biochem Mol Biol ; 147: 1-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25448746

ABSTRACT

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.


Subject(s)
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
12.
J Physiol Sci ; 64(4): 269-77, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24777837

ABSTRACT

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.


Subject(s)
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
14.
Am J Physiol Heart Circ Physiol ; 304(3): H465-72, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23203968

ABSTRACT

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.


Subject(s)
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
15.
Am J Physiol Heart Circ Physiol ; 300(5): H1661-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21335468

ABSTRACT

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.


Subject(s)
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
16.
J Appl Physiol (1985) ; 107(4): 1105-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19679745

ABSTRACT

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.


Subject(s)
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
17.
Am J Physiol Regul Integr Comp Physiol ; 292(2): R837-43, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17038443

ABSTRACT

Compared to sham-operated controls, myofilaments from hearts of ovariectomized (OVX) rats demonstrate an increase in Ca2+ sensitivity with no change in maximum tension (Wattanapermpool J and Reiser PJ. Am J Physiol 277: H467-H473, 1999). To test the significance of this modification in intact cells, we compared intracellular Ca2+ transients and shortening of ventricular myocytes isolated from sham and 10-wk OVX rats. There was a decrease in the peak Ca2+ transient with prolonged 50% decay time in OVX cardiac myocytes without changes in the resting intracellular Ca2+ concentration. Percent cell shortening was also depressed, and relaxation was prolonged in cardiac myocytes from OVX rats compared with shams. Ovariectomy induced a sensitization of the myofilaments to Ca2+. Hypercapnic acidosis suppressed the shortening of OVX myocytes to a lesser extent than that detected in shams. Moreover, a larger compensatory increase in %cell shortening was obtained in OVX myocytes during prolonged acidosis. The elevated compensation in cell shortening was related to a higher amount of increase in the amplitude of the Ca2+ transient in OVX myocytes. However, these differences in Ca2+ transients and %cell shortening were no longer evident in the presence of 1 microM cariporide, a specific inhibitor of Na+/H+ exchanger type 1 (NHE1). Our results indicate that deprivation of female sex hormones modulates the intracellular Ca2+ concentration in cardiac myocytes, possibly via an increased NHE1 activity, which may act in concert with Ca2+ hypersensitivity of myofilament activation as a determinant of sex differences in cardiac function.


Subject(s)
Acidosis, Respiratory/physiopathology , Actin Cytoskeleton/physiology , Calcium/metabolism , Gonadal Steroid Hormones/pharmacology , Hypercapnia/physiopathology , Myocytes, Cardiac/physiology , Sodium-Calcium Exchanger/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Calcium Signaling/physiology , Cell Separation , Estrogens/pharmacology , Female , Guanidines/pharmacology , Hydrogen-Ion Concentration , In Vitro Techniques , Kinetics , Muscle Fibers, Skeletal/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/drug effects , Ovariectomy , Rats , Rats, Sprague-Dawley , Sulfones/pharmacology
18.
Am J Physiol Regul Integr Comp Physiol ; 292(2): R844-51, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17038444

ABSTRACT

The amelioration of cardioprotective effect of estrogen in diabetes suggests potential interactive action of estrogen and insulin on myofilament activation. We compared Ca2+-dependent Mg2+-ATPase activity of isolated myofibrillar preparations from hearts of sham and 10-wk ovariectomized rats with or without simultaneous 8 wk-induction of diabetes and from diabetic-ovariectomized rats with estrogen and/or insulin supplementation. Similar magnitude of suppressed maximum myofibrillar ATPase activity was demonstrated in ovariectomized, diabetic, and diabetic-ovariectomized rat hearts. Such suppressed activity and the relative suppression in alpha-myosin heavy chain level in ovariectomy combined with diabetes could be completely restored by estrogen and insulin supplementation. Conversely, the myofilament Ca2+ hypersensitivity detected only in the ovariectomized but not diabetic group was also observed in diabetic-ovariectomized rats, which was restored upon estrogen supplementation. Binding kinetics of beta1-adrenergic receptors and immunoblots of beta1-adrenoceptors as well as heat shock 72 (HSP72) were analyzed to determine the association of changes in receptors and HSP72 to that of the myofilament response to Ca2+. The amount of beta1-adrenoceptors significantly increased concomitant with Ca2+ hypersensitivity of the myofilament, without differences in the receptor binding affinity among the groups. In contrast, changes in HSP72 paralleled that of maximum myofibrillar ATPase activity. These results indicate that hypersensitivity of cardiac myofilament to Ca2+ is specifically induced in ovariectomized rats even under diabetes complication and that alterations in the expression of beta1-adrenoceptors may, in part, play a mechanistic role underlying the cardioprotective effects of estrogen that act together with Ca2+ hypersensitivity of the myofilament in determining the gender difference in cardiac activation.


Subject(s)
Actin Cytoskeleton/physiology , Calcium/physiology , Diabetes Mellitus, Experimental/physiopathology , Estrogens/deficiency , Heart/physiopathology , Actomyosin/metabolism , Adenosine Triphosphatases/metabolism , Animals , Body Weight/physiology , Diabetes Mellitus, Experimental/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Female , HSP72 Heat-Shock Proteins/metabolism , Hypoglycemic Agents/pharmacology , Immunoblotting , Insulin/pharmacology , Kinetics , Myofibrils/drug effects , Myofibrils/enzymology , Myofibrils/physiology , Organ Size/physiology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/physiology , Sarcolemma/drug effects , Sarcolemma/physiology , Uterus/physiology
19.
Am J Physiol Heart Circ Physiol ; 291(3): H1101-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16517946

ABSTRACT

Alterations in the intracellular Ca2+ handling in cardiomyocytes may underlie the cardiac dysfunction observed in the ovarian sex hormone-deprived condition. To test the hypothesis that ovarian sex hormones had a significant role in the cardiac intracellular Ca2+ mobilization, the sarcoplasmic reticulum (SR) Ca2+ uptake and SR Ca2+-ATPase (SERCA) activity were determined in 10-wk ovariectomized rat hearts. With the use of left ventricular homogenate preparations, a significant suppression of maximum SR Ca2+ uptake activity, but with an increase in SR Ca2+ responsiveness, was demonstrated in ovariectomized hearts. In parallel measurements of SERCA activity in SR-enriched membrane preparations from ovariectomized hearts, a suppressed maximum SERCA activity with a leftward shift in the relationship between pCa (-log molar free Ca2+ concentration) and SERCA activity was also detected. A significant downregulation of SERCA proteins and reduction in the SERCA mRNA level were observed in association with suppressed maximum SERCA activity. While there were no changes in total phospholamban and phosphorylated Ser16 phospholamban levels, a decrease in phosphorylated Thr17 phospholamban as well as an increase in the suprainhibitory, monomeric form of phospholamban stoichiometry was found. Estrogen and progesterone supplementations were equally effective in preventing changes in ovariectomized hearts. Our data showed for the first time that female sex hormones played an important role in the regulation of the cardiac SR Ca2+ uptake. Under hormone-deficient conditions, there was an adaptive response of SERCA that escaped the regulatory effect of phospholamban.


Subject(s)
Calcium/metabolism , Estrogens/physiology , Myocytes, Cardiac/metabolism , Progesterone/physiology , Sarcoplasmic Reticulum/metabolism , Animals , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Cardiac Output, Low/metabolism , Cardiac Output, Low/physiopathology , Female , Gene Expression Regulation, Enzymologic/physiology , Myocytes, Cardiac/drug effects , Ovariectomy , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases
20.
J Appl Physiol (1985) ; 96(5): 1755-60, 2004 May.
Article in English | MEDLINE | ID: mdl-14672958

ABSTRACT

The risks associated with hormone replacement therapy, especially cardiac diseases in postmenopausal women, have prompted extensive studies for other preventive or therapeutic alternatives. We investigated the cardioprotective effects of exercise training on the changes in cardiac myofilament Ca2+ activation in 10-wk-old ovariectomized rats. The exercise groups were subjected to a 9-wk running program on a motor-driven treadmill 1 wk after surgery. The relationship between pCa (-log molar free Ca2+ concentration) and myofibrillar MgATPase activity of exercise-sham myofibrils or exercise-ovariectomized myofibrils was the same and could not be distinguished from that of sedentary-sham control hearts. In contrast, a significant suppression in maximum MgATPase activity and a leftward shift of pCa50 (half-maximally activating pCa) in the pCa-ATPase activity relationship were detected in sedentary-ovariectomized rats. Exercise training also prevented the shift in myosin heavy chain (MHC) isoforms toward beta-MHC in ovariectomized hearts. The upregulation of beta1-adrenergic receptors in the left ventricular membranes of ovariectomized rat hearts, as measured by receptor binding and immunoblot analyses, was no longer observed in exercise-ovariectomized hearts. Immunoblot analyses of heat shock protein (HSP) 72, an inducible form of HSP70, demonstrated a significant downregulation in ovariectomized hearts. Exercise training in ovariectomized rats completely reversed the expression of HSP72 to the same level as sham controls. Our results clearly indicate the cardioprotective effects of exercise training on changes in cardiac myofilament Ca2+ activation in ovariectomized rats. Alterations in expression of beta1-adrenergic receptors and HSP72 may, in part, play a mechanistic role in the cardioprotective effects.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium/metabolism , Heart/physiology , Myocardium/metabolism , Ovariectomy , Physical Conditioning, Animal/physiology , Animals , Female , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Myosin Heavy Chains/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/metabolism , Running/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...