Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Clin Sci (Lond) ; 136(21): 1537-1554, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36285636

ABSTRACT

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


Subject(s)
Cardiomyopathies , Ventricular Remodeling , Mice , Male , Animals , Isoproterenol/adverse effects , Isoproterenol/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Mice, Knockout , Cardiomyopathies/chemically induced , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/genetics , Fibrosis , Myocytes, Cardiac/metabolism , Mice, Inbred C57BL
2.
Cell Physiol Biochem ; 56(3): 293-309, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35781359

ABSTRACT

BACKGROUND/AIMS: An obesogenic diet (high fat and sugar, low fiber) is associated with an increased risk for metabolic and cardiovascular disorders. Previous studies have demonstrated that epigenetic changes can modify gene transcription and protein function, playing a key role in the development of several diseases. The methyltransferase Set7 methylates histone and non-histone proteins, influencing diverse biological and pathological processes. However, the functional role of Set7 in obesity-associated metabolic and cardiovascular complications is unknown. METHODS: Wild type and Set7 knockout female mice were fed a normal diet or an obesogenic diet for 12 weeks. Body weight gain and glucose tolerance were measured. The 3T3-L1 cells were used to determine the role of Set7 in white adipogenic differentiation. Cardiac morphology and function were evaluated by histology and echocardiography. An ex vivo Langendorff perfusion system was used to model cardiac ischemia/reperfusion (I/R). RESULTS: Here, we report that Set7 protein levels were enhanced in the heart and perigonadal adipose tissue (PAT) of female mice fed an obesogenic diet. Significantly, loss of Set7 prevented obesogenic diet-induced glucose intolerance in female mice although it did not affect the obesogenic diet-induced increase in body weight gain and adiposity in these animals, nor did Set7 inhibition change white adipogenic differentiation in vitro. In addition, loss of Set7 prevented the compromised cardiac functional recovery following ischemia and reperfusion (I/R) injury in obesogenic diet-fed female mice; however, deletion of Set7 did not influence obesogenic diet-induced cardiac hypertrophy nor the hemodynamic and echocardiographic parameters. CONCLUSION: These data indicate that Set7 plays a key role in obesogenic diet-induced glucose intolerance and compromised myocardial functional recovery after I/R in obese female mice.


Subject(s)
Glucose Intolerance , Animals , Diet, High-Fat/adverse effects , Female , Ischemia , Mice , Mice, Knockout , Mice, Obese , Obesity/metabolism , Reperfusion/adverse effects
3.
Int J Mol Sci ; 23(12)2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35742928

ABSTRACT

Leptin resistance is a hallmark of obesity. Treatments aiming to improve leptin sensitivity are considered a promising therapeutical approach against obesity. However, leptin receptor (LepR) signaling also modulates several neurovegetative aspects, such as the cardiovascular system and hepatic gluconeogenesis. Thus, we investigated the long-term consequences of increased leptin sensitivity, considering the potential beneficial and deleterious effects. To generate a mouse model with increased leptin sensitivity, the suppressor of cytokine signaling 3 (SOCS3) was ablated in LepR-expressing cells (LepR∆SOCS3 mice). LepR∆SOCS3 mice displayed reduced food intake, body adiposity and weight gain, as well as improved glucose tolerance and insulin sensitivity, and were protected against aging-induced leptin resistance. Surprisingly, a very high mortality rate was observed in aging LepR∆SOCS3 mice. LepR∆SOCS3 mice showed cardiomyocyte hypertrophy, increased myocardial fibrosis and reduced cardiovascular capacity. LepR∆SOCS3 mice exhibited impaired post-ischemic cardiac functional recovery and middle-aged LepR∆SOCS3 mice showed substantial arhythmic events during the post-ischemic reperfusion period. Finally, LepR∆SOCS3 mice exhibited fasting-induced hypoglycemia and impaired counterregulatory response to glucopenia associated with reduced gluconeogenesis. In conclusion, although increased sensitivity to leptin improved the energy and glucose homeostasis of aging LepR∆SOCS3 mice, major autonomic/neurovegetative dysfunctions compromised the health and longevity of these animals. Consequently, these potentially negative aspects need to be considered in the therapies that increase leptin sensitivity chronically.


Subject(s)
Heart Diseases , Receptors, Leptin , Animals , Energy Metabolism , Glucose/metabolism , Heart Diseases/metabolism , Leptin/metabolism , Mice , Neurons/metabolism , Obesity/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism
4.
Exp Physiol ; 107(8): 892-905, 2022 08.
Article in English | MEDLINE | ID: mdl-35765992

ABSTRACT

NEW FINDINGS: What is the central question of this study? What is the effect of an obesogenic diet on the expression of microRNAs (miRNAs) involved in cardiac hypertrophy in female mice? What is the main finding and its importance? Female mice fed an obesogenic diet exhibited cardiac hypertrophy associated with increased levels of miRNA-143-3p, decreased mRNA levels of Sox6 and increased mRNA levels of Myh7. Inhibition of miRNA-143-3p increased Sox6 mRNA levels and reduced Myh7 expression in cardiomyocytes, and prevented angiotensin II-induced cardiomyocyte hypertrophy. The results indicate that the miRNA-143-3p-Sox6-Myh7 pathway may play a key role in obesity-induced cardiac hypertrophy. ABSTRACT: Obesity induces cardiometabolic disorders associated with a high risk of mortality. We have previously shown that the microRNA (miRNA) expression profile is changed in obesity-induced cardiac hypertrophy in male mice. Here, we investigated the effect of an obesogenic diet on the expression of miRNAs involved in cardiac hypertrophy in female mice. Female mice fed an obesogenic diet displayed an increased body weight gain, glucose intolerance, insulin resistance and dyslipidaemia. In addition, obese female mice exhibited cardiac hypertrophy associated with increased levels of several miRNAs, including miR-143-3p. Bioinformatic analysis identified Sox6, regulator of Myh7 gene transcription, as a predicted target of miR-143-3p. Female mice fed an obesogenic diet exhibited decreased mRNA levels of Sox6 and increased expression of Myh7 in the heart. Loss-of-function studies in cardiomyocytes revealed that inhibition of miR-143-3p increased Sox6 mRNA levels and reduced Myh7 expression. Collectively, our results indicate that obesity-associated cardiac hypertrophy in female mice is accompanied by alterations in diverse miRNAs, and suggest that the miR-143-3p-Sox6-Myh7 pathway may play a key role in obesity-induced cardiac hypertrophy.


Subject(s)
Cardiomegaly , MicroRNAs , Animals , Cardiomegaly/metabolism , Diet , Female , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/metabolism , Obesity/metabolism , RNA, Messenger/metabolism , SOXD Transcription Factors/metabolism
5.
J Cell Physiol ; 236(4): 3059-3072, 2021 04.
Article in English | MEDLINE | ID: mdl-32964425

ABSTRACT

Clinical studies have shown a correlation between thyroid disorders and cardiac diseases. High levels of triiodothyronine (T3) induce cardiac hypertrophy, a risk factor for cardiac complications and heart failure. Previous results have demonstrated that angiotensin-(1-7) is able to block T3-induced cardiac hypertrophy; however, the molecular mechanisms involved in this event have not been fully elucidated. Here, we evidenced the contribution of FOXO3 signaling to angiotensin-(1-7) effects. Angiotensin-(1-7) treatment increased nuclear FOXO3 levels and reduced p-FOXO3 levels (inactive form) in isolated cardiomyocytes. Knockdown of FOXO3 by RNA silencing abrogated the antihypertrophic effect of angiotensin-(1-7). Increased expression of antioxidant enzymes superoxide dismutase 1 (SOD1 and catalase) and lower levels of reactive oxygen species and nuclear factor-κB (NF-κB) were observed after angiotensin-(1-7) treatment in vitro. Consistent with these results, transgenic rats overexpressing angiotensin-(1-7) displayed increased nuclear FOXO3 and SOD1 levels and reduced NF-κB levels in the heart. These results provide a new molecular mechanism responsible for the antihypertrophic effect of angiotensin-(1-7), which may contribute to future therapeutic targets.


Subject(s)
Angiotensin I/pharmacology , Catalase/metabolism , Forkhead Box Protein O3/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Peptide Fragments/pharmacology , Superoxide Dismutase-1/metabolism , Triiodothyronine/adverse effects , Up-Regulation , Animals , Antioxidants/metabolism , Down-Regulation/drug effects , Hypertrophy , Male , Models, Biological , Myocytes, Cardiac/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/drug effects
6.
Cell Physiol Biochem ; 54(6): 1199-1217, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33252886

ABSTRACT

BACKGROUND/AIMS: Obesity is a risk factor associated with cardiometabolic complications. Recently, we reported that miRNA-22 deletion attenuated high-fat diet-induced adiposity and prevented dyslipidemia without affecting cardiac hypertrophy in male mice. In this study, we examined the impact of miRNA-22 in obesogenic diet-induced cardiovascular and metabolic disorders in females. METHODS: Wild type (WT) and miRNA-22 knockout (miRNA-22 KO) females were fed a control or an obesogenic diet. Body weight gain, adiposity, glucose tolerance, insulin tolerance, and plasma levels of total cholesterol and triglycerides were measured. Cardiac and white adipose tissue remodeling was assessed by histological analyses. Echocardiography was used to evaluate cardiac function and morphology. RNA-sequencing analysis was employed to characterize mRNA expression profiles in female hearts. RESULTS: Loss of miRNA-22 attenuated body weight gain, adiposity, and prevented obesogenic diet-induced insulin resistance and dyslipidemia in females. WT obese females developed cardiac hypertrophy. Interestingly, miRNA-22 KO females displayed cardiac hypertrophy without left ventricular dysfunction and myocardial fibrosis. Both miRNA-22 deletion and obesogenic diet changed mRNA expression profiles in female hearts. Enrichment analysis revealed that genes associated with regulation of the force of heart contraction, protein folding and fatty acid oxidation were enriched in hearts of WT obese females. In addition, genes related to thyroid hormone responses, heart growth and PI3K signaling were enriched in hearts of miRNA-22 KO females. Interestingly, miRNA-22 KO obese females exhibited reduced mRNA levels of Yap1, Egfr and Tgfbr1 compared to their respective controls. CONCLUSION: This study reveals that miRNA-22 deletion induces cardiac hypertrophy in females without affecting myocardial function. In addition, our findings suggest miRNA-22 as a potential therapeutic target to treat obesity-related metabolic disorders in females.


Subject(s)
Cardiomegaly , Diet, High-Fat/adverse effects , Gene Deletion , Metabolic Diseases , MicroRNAs/genetics , Myocardium , Obesity , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Female , Metabolic Diseases/chemically induced , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Mice, Knockout , MicroRNAs/metabolism , Myocardium/metabolism , Myocardium/pathology , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Obesity/pathology
7.
Mol Cell Endocrinol ; 518: 110972, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32777452

ABSTRACT

Although myocardial growth corresponds to an adaptive response to maintain cardiac contractile function, the cardiac hypertrophy is a condition that occurs in many cardiovascular diseases and typically precedes the onset of heart failure. Different endocrine factors such as thyroid hormones, insulin, insulin-like growth factor 1 (IGF-1), angiotensin II (Ang II), endothelin (ET-1), catecholamines, estrogen, among others represent important stimuli to cardiomyocyte hypertrophy. Thus, numerous endocrine disorders manifested as changes in the local environment or multiple organ systems are especially important in the context of progression from cardiac hypertrophy to heart failure. Based on that information, this review summarizes experimental findings regarding the influence of such hormones upon signalling pathways associated with cardiac hypertrophy. Understanding mechanisms through which hormones differentially regulate cardiac hypertrophy could open ways to obtain therapeutic approaches that contribute to prevent or delay the onset of heart failure related to endocrine diseases.


Subject(s)
Cardiomegaly/metabolism , Endocrine System/metabolism , Signal Transduction , Angiotensin II/metabolism , Animals , Disease Progression , Humans , Insulin/metabolism , Thyroid Hormones/metabolism
8.
Mol Cell Endocrinol ; 498: 110576, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31520674

ABSTRACT

Obesity is the major risk factor for several cardiovascular and metabolic disorders. Previous studies reported that deletion of Angiotensin II type 2 receptor (AT2R) protects against metabolic dysfunctions induced by high fat (HF) diet. However, the role of AT2R in obesity-induced cardiac hypertrophy remains unclear. Male AT2R knockout (AT2RKO) and wild type (AT2RWT) mice were fed with control or HF diet for 10 weeks. HF diet increased cardiac expression of AT2R in obese mice. Deletion of AT2R did not affect body weight gain, glucose intolerance and fat mass gain induced by HF feeding. However, loss of AT2R prevented HF diet-induced hypercholesterolemia and cardiac remodeling. Mechanistically, we found that pharmacological inhibition or knockdown of AT2R prevented leptin-induced cardiomyocyte hypertrophy in vitro. Collectively, our results suggest that AT2R is involved in obesity-induced cardiac hypertrophy.


Subject(s)
Cardiomegaly/etiology , Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Hypercholesterolemia/etiology , Insulin Resistance , Obesity/complications , Receptor, Angiotensin, Type 2/physiology , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Leptin/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
9.
J Cell Physiol ; 234(6): 9399-9407, 2019 06.
Article in English | MEDLINE | ID: mdl-30362585

ABSTRACT

Several studies have shown the role of microRNAs (miRNAs) in myocardial dysfunction in response to ischemia/reperfusion (I/R). In this study, we investigated the impact of high fat (HF) diet in the myocardial susceptibility to I/R injury, as well as in the expression of miRNA-29b. Isolated heart experiments using the ex vivo Langendorff perfusion model were used to induce cardiac I/R injury. HF diet-induced cardiac hypertrophy and impaired cardiac functional recovery after I/R. miRNA-29b, which targets Col1, was reduced in the heart of HF diet-fed mice, whereas the cardiac expression of Col1 was increased. In addition, hypoxia-reoxygenation (H/R) reduced the expression of miRNA-29b in cardiomyoblasts cultures. However, the overexpression of miRNA-29b in cardiomyoblasts reduced p53 mRNA levels and H/R injury, suggesting that downregulation of miRNA-29b may be involved in I/R injury. Together, our findings suggest that the reduced expression of miRNA-29b may be involved in the deteriorated cardiac functional recovery following I/R in obese mice.


Subject(s)
Diet, High-Fat , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocardium/metabolism , Myocardium/pathology , Animals , Body Weight , Cell Line , Collagen/genetics , Collagen/metabolism , Dyslipidemias/complications , Dyslipidemias/pathology , Glucose Intolerance/complications , Glucose Intolerance/pathology , Heart Function Tests , Hypoxia/complications , Hypoxia/genetics , Hypoxia/pathology , L-Lactate Dehydrogenase/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocardial Reperfusion Injury/complications , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology
10.
Clin Sci (Lond) ; 132(11): 1117-1133, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29685981

ABSTRACT

Patients with hyperthyroidism exhibit increased risk of development and progression of cardiac diseases. The activation of the renin-angiotensin system (RAS) has been indirectly implicated in these cardiac effects observed in hyperthyroidism. Angiotensin-(1-7) (Ang-(1-7)) has previously been shown to counterbalance pathological effects of angiotensin II (Ang II). The aim of the present study was to investigate the effects of elevated circulating Ang-(1-7) levels on cardiac effects promoted by hyperthyroidism in a transgenic rat (TG) model that constitutively overexpresses an Ang-(1-7)-producing fusion protein [TGR(A1-7)3292]. TG and wild-type (WT) rats received daily injections (i.p.) of triiodothyronine (T3; 7 µg/100 g of body weight (BW)) or vehicle for 14 days. In contrast with WT rats, the TG rats did not develop cardiac hypertrophy after T3 treatment. Indeed, TG rats displayed reduced systolic blood pressure (SBP) and cardiac hyperdynamic condition induced by hyperthyroidism. Moreover, increased plasma levels of Ang II observed in hyperthyroid WT rats were prevented in TG rats. TG rats were protected from glycogen synthase kinase 3ß (GSK3ß) inactivation and nuclear factor of activated T cells (NFAT) nuclear accumulation induced by T3. In vitro studies evidenced that Ang-(1-7) prevented cardiomyocyte hypertrophy and GSK3ß inactivation induced by T3. Taken together, these data reveal an important cardioprotective action of Ang-(1-7) in experimental model of hyperthyroidism.


Subject(s)
Angiotensin I/physiology , Cardiomegaly/etiology , Glycogen Synthase Kinase 3 beta/physiology , Hyperthyroidism/complications , NFATC Transcription Factors/physiology , Peptide Fragments/physiology , Angiotensin I/blood , Angiotensin I/pharmacology , Animals , Cardiomegaly/blood , Cardiomegaly/diagnostic imaging , Cells, Cultured , Echocardiography , Hyperthyroidism/blood , Hyperthyroidism/chemically induced , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Peptide Fragments/blood , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Triiodothyronine/pharmacology
11.
Heart Vessels ; 33(6): 671-681, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29218410

ABSTRACT

Studies have demonstrated that thyroid hormone (T3) can precondition the heart against ischaemic injury and improve post-ischaemic recovery. This study investigated whether the AT2 receptor (AT2R) is involved in cardioprotection and the potential molecular mechanism responsible for this effect. Hyperthyroidism was induced in male wild-type (WT) and AT2R knockout (KO) mice by administering daily intraperitoneal injections of T3 (7 µg/100 g body weight) for 14 days. The mouse hearts were harvested and perfused with a Krebs-Henseleit solution at a constant flow in a Langendorff set-up. After 30 min of stabilization, the hearts were subjected to global ischaemia for 20 min and reperfused for 45 min. Baseline cardiac function was assessed by measuring four parameters: LVDP (mmHg), heart rate (bpm), + dP/dt and - dP/dt (mmHg/s). After reperfusion, the total protein from cardiac ventricles was obtained, and the Akt signalling pathway and NO production were evaluated. Post-ischaemic functional recovery was significantly greater (p < 0.05) in the T3-treated WT mice compared to the control, demonstrating the cardioprotective effect of T3. This effect was abolished in T3-treated KO mice, demonstrating the physiological relevance of AT2R to the cardioprotective phenotype induced by T3. Akt activation, iNOS expression and NO production increased in cardiac tissue after T3 treatment in the WT animals, but no difference was observed after treatment in the KO mice. This study indicates that AT2R acts as a cardioprotector in the case of hyperthyroidism. Strategies targeting AT2R agonists might improve cardiac function through NO production and suggest potential therapeutic targets for heart diseases.


Subject(s)
Coronary Circulation/ethics , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Nitric Oxide/biosynthesis , Receptor, Angiotensin, Type 2/metabolism , Thyroid Hormones/pharmacology , Animals , Coronary Circulation/drug effects , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology
12.
J Cell Physiol ; 231(8): 1771-83, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26638879

ABSTRACT

Recent studies have revealed the involvement of microRNAs (miRNAs) in the control of cardiac hypertrophy and myocardial function. In addition, several reports have demonstrated that high fat (HF) diet induces cardiac hypertrophy and remodeling. In the current study, we investigated the effect of diets containing different percentages of fat on the cardiac miRNA expression signature. To address this question, male C57Bl/6 mice were fed with a low fat (LF) diet or two HF diets, containing 45 kcal% fat (HF45%) and 60 kcal% fat (HF60%) for 10 and 20 weeks. HF60% diet promoted an increase on body weight, fasting glycemia, insulin, leptin, total cholesterol, triglycerides, and induced glucose intolerance. HF feeding promoted cardiac remodeling, as evidenced by increased cardiomyocyte transverse diameter and interstitial fibrosis. RNA sequencing analysis demonstrated that HF feeding induced distinct miRNA expression patterns in the heart. HF45% diet for 10 and 20 weeks changed the abundance of 64 and 26 miRNAs in the heart, respectively. On the other hand, HF60% diet for 10 and 20 weeks altered the abundance of 27 and 88 miRNAs in the heart, respectively. Bioinformatics analysis indicated that insulin signaling pathway was overrepresented in response to HF diet. An inverse correlation was observed between cardiac levels of GLUT4 and miRNA-29c. Similarly, we found an inverse correlation between expression of GSK3ß and the expression of miRNA-21a-3p, miRNA-29c-3p, miRNA-144-3p, and miRNA-195a-3p. In addition, miRNA-1 overexpression prevented cardiomyocyte hypertrophy. Taken together, our results revealed differentially expressed miRNA signatures in the heart in response to different HF diets. J. Cell. Physiol. 231: 1771-1783, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cardiomegaly/genetics , Diet, High-Fat , Gene Expression Profiling , MicroRNAs/genetics , Myocytes, Cardiac , Ventricular Remodeling/genetics , Animals , Animals, Newborn , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Cells, Cultured , Computational Biology , Diet, Fat-Restricted , Disease Models, Animal , Dyslipidemias/genetics , Dyslipidemias/metabolism , Fibrosis , Gene Expression Profiling/methods , Gene Expression Regulation , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin/genetics , Insulin/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Signal Transduction/genetics , Time Factors
13.
Ther Adv Cardiovasc Dis ; 10(4): 192-202, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26715125

ABSTRACT

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


Subject(s)
Angiotensin I/analysis , Cardiomegaly/etiology , Hyperthyroidism/complications , Myocardium/metabolism , Peptide Fragments/analysis , Peptidyl-Dipeptidase A/analysis , Proto-Oncogene Proteins/analysis , Receptors, G-Protein-Coupled/analysis , Angiotensin I/physiology , Angiotensin-Converting Enzyme 2 , Animals , Male , Peptide Fragments/physiology , Peptidyl-Dipeptidase A/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins/physiology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/physiology , Renin-Angiotensin System/physiology
14.
Mol Cell Endocrinol ; 416: 1-8, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26277399

ABSTRACT

Suboptimal intrauterine conditions as changed hormone levels during critical periods of the development are considered an insult and implicate in physiological adaptations which may result in pathological outcomes in later life. This study evaluated the effect of maternal hyperthyroidism (hyper) on cardiac function in adult offspring and the possible involvement of cardiac Renin-Angiotensin System (RAS) in this process. Wistar dams received orally thyroxin (12 mg/L) from gestational day 9 (GD9) until GD18. Adult offspring at postnatal day 90 (PND90) from hyper dams presented increased SBP evaluated by plethysmography and worse recovery after ischemia-reperfusion (I/R), as evidenced by decreased LVDP, +dP/dT and -dP/dT at 25 min of reperfusion and by increased infarct size. Increased cardiac Angiotensin I/II levels and AT1R in hyper offspring were verified. Herein, we provide evidences that maternal hyperthyroidism leads to altered expression of RAS components in adult offspring, which may be correlated with worse recovery of the cardiac performance after ischemic insults and hypertension.


Subject(s)
Hypertension/etiology , Hyperthyroidism/metabolism , Myocardial Reperfusion Injury/etiology , Pregnancy Complications/metabolism , Renin-Angiotensin System , Thyroxine/metabolism , Angiotensin I/metabolism , Angiotensin II/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Disease Susceptibility , Female , Male , Plethysmography , Pregnancy , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Thyroxine/adverse effects
15.
J Renin Angiotensin Aldosterone Syst ; 15(1): 52-60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23257210

ABSTRACT

INTRODUCTION: Changes in perinatal environment can lead to physiological, morphological, or metabolic alterations in adult life. It is well known that thyroid hormones (TH) are critical for the development, growth, and maturation of organs and systems. In addition, TH interact with the renin-angiotensin system (RAS), and both play a critical role in adult cardiovascular function. The objective of this study was to evaluate the effect of maternal hyperthyroidism on cardiac RAS components in pups during development. MATERIALS AND METHODS: From gestational day nine (GD9), pregnant Wistar rats received thyroxine (T4, 12 mg/l in tap water; Hyper group) or vehicle (control group). Dams and pups were killed on GD18 and GD20. RESULTS: Serum concentrations of triiodothyronine (T3) and T4 were higher in the Hyper group than in the control group dams. Cardiac hypertrophy was observed in Hyper pups on GD20. Cardiac angiotensin-converting enzyme (ACE) activity was significantly lower in Hyper pups on both GD18 and GD20, but there was no difference in Ang I/Ang II levels. Ang II receptors expression was higher in the Hyper pup heart on GD18. CONCLUSIONS: Maternal hyperthyroidism is associated with alterations in fetal development and altered pattern of expression in RAS components, which in addition to cardiac hypertrophy observed on GD20 may represent an important predisposing factor to cardiovascular diseases in adult life.


Subject(s)
Cardiomegaly/chemically induced , Hyperthyroidism/physiopathology , Mothers , Renin-Angiotensin System/drug effects , Thyroxine/pharmacology , Animals , Animals, Newborn , Female , Fetal Development/drug effects , Myocardium/metabolism , Peptidyl-Dipeptidase A/metabolism , Pregnancy , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 2/biosynthesis , Renin-Angiotensin System/physiology , Thyroxine/blood , Triiodothyronine/blood , Up-Regulation
16.
Mol Endocrinol ; 27(12): 2055-65, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24176915

ABSTRACT

Although it is well known that the thyroid hormone (T3) is an important positive regulator of cardiac function over a short term and that it also promotes deleterious effects over a long term, the molecular mechanisms for such effects are not yet well understood. Because most alterations in cardiac function are associated with changes in sarcomeric machinery, the present work was undertaken to find novel sarcomeric hot spots driven by T3 in the heart. A microarray analysis indicated that the M-band is a major hot spot, and the structural sarcomeric gene coding for the M-protein is severely down-regulated by T3. Real-time quantitative PCR-based measurements confirmed that T3 (1, 5, 50, and 100 physiological doses for 2 days) sharply decreased the M-protein gene and protein expression in vivo in a dose-dependent manner. Furthermore, the M-protein gene expression was elevated 3.4-fold in hypothyroid rats. Accordingly, T3 was able to rapidly and strongly reduce the M-protein gene expression in neonatal cardiomyocytes. Deletions at the M-protein promoter and bioinformatics approach suggested an area responsive to T3, which was confirmed by chromatin immunoprecipitation assay. Functional assays in cultured neonatal cardiomyocytes revealed that depletion of M-protein (by small interfering RNA) drives a severe decrease in speed of contraction. Interestingly, mRNA and protein levels of other M-band components, myomesin and embryonic-heart myomesin, were not altered by T3. We concluded that the M-protein expression is strongly and rapidly repressed by T3 in cardiomyocytes, which represents an important aspect for the basis of T3-dependent sarcomeric deleterious effects in the heart.


Subject(s)
Cardiomegaly/genetics , Connectin/genetics , Down-Regulation/genetics , Thyroid Hormones/pharmacology , Animals , Animals, Newborn , Base Sequence , Cardiomegaly/etiology , Cardiomegaly/physiopathology , Cell Line , Cells, Cultured , Connectin/metabolism , Down-Regulation/drug effects , Hypothyroidism/genetics , Male , Mice , Molecular Sequence Data , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Thyroid Hormone/metabolism , Sarcomeres/metabolism , Thyrotoxicosis/complications , Triiodothyronine/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
17.
PLoS One ; 8(4): e61982, 2013.
Article in English | MEDLINE | ID: mdl-23637941

ABSTRACT

Hyperthyroidism is characterized by increased vascular relaxation and decreased vascular contraction and is associated with augmented levels of triiodothyronine (T3) that contribute to the diminished systemic vascular resistance found in this condition. T3 leads to augmented NO production via PI3K/Akt signaling pathway, which in turn causes vascular smooth muscle cell (VSMC) relaxation; however, the underlying mechanisms involved remain largely unknown. Evidence from human and animal studies demonstrates that the renin-angiotensin system (RAS) plays a crucial role in vascular function and also mediates some of cardiovascular effects found during hyperthyroidism. Thus, in this study, we hypothesized that type 2 angiotensin II receptor (AT2R), a key component of RAS vasodilatory actions, mediates T3 induced-decreased vascular contraction. Marked induction of AT2R expression was observed in aortas from T3-induced hyperthyroid rats (Hyper). These vessels showed decreased protein levels of the contractile apparatus: α-actin, calponin and phosphorylated myosin light chain (p-MLC). Vascular reactivity studies showed that denuded aortic rings from Hyper rats exhibited decreased maximal contractile response to angiotensin II (AngII), which was attenuated in aortic rings pre-incubated with an AT2R blocker. Further study showed that cultured VSMC stimulated with T3 (0.1 µmol/L) for 24 hours had increased AT2R gene and protein expression. Augmented NO levels and decreased p-MLC levels were found in VSMC stimulated with T3, both of which were reversed by a PI3K/Akt inhibitor and AT2R blocker. These findings indicate for the first time that the AT2R/Akt/NO pathway contributes to decreased contractile responses in rat aorta, promoted by T3, and this mechanism is independent from the endothelium.


Subject(s)
Hyperthyroidism/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction , Angiotensin II/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Hyperthyroidism/genetics , Male , Models, Biological , Myocytes, Smooth Muscle/drug effects , Rats , Receptor, Angiotensin, Type 2/genetics , Signal Transduction/drug effects , Triiodothyronine/pharmacology , Vasoconstriction/drug effects
18.
Life Sci ; 91(3-4): 137-42, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22771697

ABSTRACT

AIMS: To investigate the role of TH (thyroid hormones) in 5'-nucleotidase activity and expression in cardiac soluble fraction (SF). MAIN METHODS: Male Wistar rats received daily injections of T4 (10, 25 or 50 µg T4/100g body weight) for 14 days to develop a hyperthyroidism condition. Thyroidectomy was performed in other animals to mimic hypothyroidism, and 14 days after surgery they were submitted to TH replacement therapy. KEY FINDINGS: T4 reduced the 5'-nucleotidase activity (T4-25, P<0.05 and T4-50, P<0.01) in the SF. Conversely, hypothyroidism significantly increased the 5'-nucleotidase activity in this fraction (P<0.001) and TH replacement therapy reversed the latter result (P<0.001 compared to hypothyroid group). The analysis of protein expression in the SF showed that 5'-nucleotidase was more expressed in hypothyroid than in the control group and that the phosphorylated state of PKC observed in this condition may contribute to a possible mechanism of 5'-nucleotidase modulation by thyroid status. SIGNIFICANCE: Taken together, these data reveal that TH can influence adenosine production by modulating 5'-nucleotidase activity and expression, which may contribute to the cardioprotective effect and the maintenance of cardiac function under TH privation.


Subject(s)
5'-Nucleotidase/metabolism , Thyroid Hormones/metabolism , Animals , Blood Pressure , Body Weight , Gene Expression Regulation , Hemodynamics , Hyperthyroidism/metabolism , Hypothyroidism/metabolism , Male , Models, Biological , Myocardium/metabolism , Nucleotides/chemistry , Protein Kinase C/metabolism , Rats , Rats, Wistar , Solubility , Subcellular Fractions/metabolism
19.
Biofactors ; 37(1): 40-5, 2011.
Article in English | MEDLINE | ID: mdl-21328626

ABSTRACT

The effects of ATP, ADP, and adenosine in the processes of platelet aggregation, vasodilatation, and coronary flow have been known for many years. The sequential hydrolysis of ATP to adenosine by soluble nucleotidases constitutes the main system for rapid inactivation of circulating adenine nucleotides. Thyroid disorders affect a number of biological factors including adenosine levels in different fractions. Then, we intend to investigate if the soluble nucleotidases responsible for the ATP, ADP, and AMP hydrolysis are affected by variations in the thyroid hormone levels in blood serum from adult rats. Hyperthyroidism was induced by daily intraperitoneal injections of L-thyroxine (T4) (2.5 and 10.0 µg/100 g body weight, respectively) for 7 or 14 days. Hypothyroidism was induced by thyroidectomy and methimazole (0.05%) added to their drinking water during 7 or 14 days. The treatments efficacy was confirmed by determination of hemodynamic parameters and cardiac hypertrophy evaluation. T4 treatment predominantly inhibited, and hypothyroidism (14 days after thyroidectomy) predominantly increased the ATP, ADP, and AMP hydrolysis in rat blood serum. These results suggest that both excess and deficiency of thyroid hormones can modulate the ATP diphosphohydrolase and 5'-nucleotidase activities in rat blood serum and consequently modulate the effects mediated by these enzymes and their products in vascular system.


Subject(s)
Adenine Nucleotides/blood , Thyroxine/physiology , Animals , Antithyroid Agents , Enzyme Assays , Hemodynamics , Hydrolysis , Hyperthyroidism/blood , Hyperthyroidism/chemically induced , Hypothyroidism/blood , Hypothyroidism/etiology , Male , Methimazole , Rats , Rats, Wistar , Thyroidectomy , Thyroxine/deficiency
20.
Cardiovasc Res ; 85(3): 560-70, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19734167

ABSTRACT

AIMS: Thyroid hormone (TH) rapidly relaxes vascular smooth muscle cells (VSMCs). However, the mechanisms involved in this effect remain unclear. We hypothesize that TH-induced rapid vascular relaxation is mediated by VSMC-derived nitric oxide (NO) production and is associated with the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) signalling pathway. METHODS AND RESULTS: NO levels were determined using a NO-specific fluorescent dye (DAF-2) and nitrite (NO2-) levels. Expression of NO synthase (NOS) isoforms and proteins of the PI3K/Akt pathway was determined by both western blotting and immunocytochemistry. Myosin light chain (MLC) phosphorylation levels were also investigated by western blotting. Exposure of cultured VSMCs from rat thoracic aortas to triiodothyronine (T3) resulted in a significant decrease of MLC phosphorylation levels. T3 also induced a rapid increase in Akt phosphorylation and increased NO production in a dose-dependent manner (0.001-1 microM). VSMCs stimulated with T3 for 30 min showed an increase in the expression of all three NOS isoforms and augmented NO production, effects that were prevented by inhibitors of PI3K. Vascular reactivity studies showed that vessels treated with T3 displayed a decreased response to phenylephrine, which was reversed by NOS inhibition. These data suggest that T3 treatment induces greater generation of NO both in aorta and VSMCs and that this phenomenon is endothelium independent. In addition, these findings show for the first time that the PI3K/Akt signalling pathway is involved in T3-induced NO production by VSMCs, which occurs with expressive participation of inducible and neuronal NOS. CONCLUSION: Our data strongly indicate that T3 causes NO-dependent rapid relaxation of VSMC and that this effect is mediated by the PI3K/Akt signalling pathway.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/biosynthesis , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects , Thyroid Hormones/pharmacology , Animals , Cells, Cultured , Endothelium, Vascular/physiology , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/metabolism , Nitric Oxide Synthase/physiology , Phenylephrine/pharmacology , Phosphorylation , Rats , Rats, Wistar , Vasoconstriction
SELECTION OF CITATIONS
SEARCH DETAIL
...