Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Physiol Rep ; 11(24): e15866, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38114067

ABSTRACT

Human bone mesenchymal stem cell-derived extracellular vesicles (HBMSC-EV) have been used successfully in animal models of myocardial ischemia, yet have dampened effects in metabolic syndrome through unknown mechanisms. This study demonstrates the basal differences between non-diabetic human coronary artery endothelial cells (HCAEC) and diabetic HCAEC (DM-HCAEC), and how these cells respond to the treatment of HBMSC-EV. HCAEC and DM-HCAEC were treated with HBMSC-EV for 6 h. Proteomics, western blot analysis, and tube formation assays were performed. Key metabolic, growth, and stress/starvation cellular responses were significantly altered in DM-HCAEC in comparison to that of HCAEC at baseline. Proteomics demonstrated increased phosphorus metabolic process and immune pathways and decreased RNA processing and biosynthetic pathways in DM-HCAEC. Similar to previous in vivo findings, HCAEC responded to the HBMSC-EV with regenerative and anti-inflammatory effects through the upregulation of multiple RNA pathways and downregulation of immune cell activation pathways. In contrast, DM-HCAEC had a significantly diminished response to HBMSC-EV, likely due to the baseline abnormalities in DM-HCAEC. To achieve the full benefits of HBMSC-EV and for a successful transition of this potential therapeutic agent to clinical studies, the abnormalities found in DM-HCAEC will need to be further studied.


Subject(s)
Diabetes Mellitus , Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Humans , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Diabetes Mellitus/metabolism
2.
Can J Physiol Pharmacol ; 101(9): 447-454, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37581356

ABSTRACT

Oxidative stress is involved in increased pulmonary vascular resistance (PVR) and right ventricular (RV) hypertrophy, characteristics of pulmonary arterial hypertension (PAH). Copaiba oil, an antioxidant compound, could attenuate PAH damage. This study's aim was to determine the effects of copaiba oil on lung oxidative stress, PVR, and mean pulmonary arterial pressure (mPAP) in the monocrotaline (MCT) model of PAH. Male Wistar rats (170 g, n = 7/group) were divided into four groups: control, MCT, copaiba oil, and MCT + copaiba oil (MCT-O). PAH was induced by MCT (60 mg/kg i.p.) and, after 1 week, the treatment with copaiba oil (400 mg/kg/day gavage) was started for 14 days. Echocardiographic and hemodynamic measurements were performed. RV was collected for morphometric evaluations and lungs and the pulmonary artery were used for biochemical analysis. Copaiba oil significantly reduced RV hypertrophy, PVR, mPAP, and antioxidant enzyme activities in the MCT-O group. Moreover, increased nitric oxide synthase and decreased NADPH oxidase activities were observed in the MCT-O group. In conclusion, copaiba oil was able to improve the balance between nitric oxide and reactive oxygen species in lungs and the pulmonary artery and to reduce PVR, which could explain a decrease in RV hypertrophy in this PAH model.


Subject(s)
Hypertension, Pulmonary , Oils, Volatile , Pulmonary Arterial Hypertension , Rats , Male , Animals , Rats, Wistar , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Monocrotaline/adverse effects , Nitric Oxide , Antioxidants/pharmacology , Biological Availability , Lung , Pulmonary Artery , Familial Primary Pulmonary Hypertension , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Oils, Volatile/pharmacology , Disease Models, Animal
3.
Physiol Rep ; 11(6): e15568, 2023 03.
Article in English | MEDLINE | ID: mdl-36967241

ABSTRACT

In animal models, human bone marrow mesenchymal stem cell-derived extracellular vesicles (MSC-EV) have been found to have beneficial effects in cardiovascular disease, but only when administered via intramyocardial injection. The biodistribution of either intravenous or intramyocardial injection of MSC-EV in the presence of myocardial injury is uncharacterized at this time. We hypothesized that intramyocardial injection will ensure delivery of MSC-EV to the ischemic myocardium, while intravenous injection will not. Human bone marrow mesenchymal stem cells were cultured and the MSC-EV were isolated and characterized. The MSC-EVs were then labeled with DiD lipid dye. FVB mice with normal cardiac function underwent left coronary artery ligation followed by either peri-infarct intramyocardial or tail vein injection of 3*106 or 2*109 particles of DiD-labeled MSC-EV or a DiD-saline control. The heart, lungs, liver, spleen and kidneys were harvested 2 h post-injection and were submitted for fluorescent molecular tomography imaging. Myocardial uptake of MSC-EV was only visualized after intramyocardial injection of 2*109 MSC-EV particles (p = 0.01) compared to control, and there were no differences in cardiac fluorescence after tail vein injection of MSC-EV (p = 0.5). There was no significantly detectable MSC-EV uptake in other organs after intramyocardial injection. After tail vein injection of 2*109 particles of MSC-EV, the liver (p = 0.02) and spleen (p = 0.04) appeared to have diffuse MSC-EV uptake compared to controls. Even in the presence of myocardial injury, only intramyocardial but not intravenous administration resulted in detectable levels of MSC-EV in the ischemic myocardium. This study confirms the role for intramyocardial injection in maximal and effective delivery of MSC-EV. Our ongoing studies aimed at developing bioengineered MSC-EV for targeted delivery to the heart may render MSC-EV clinically applicable for cardiovascular disease.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Myocardial Infarction , Mice , Animals , Humans , Injections, Intravenous , Tissue Distribution , Extracellular Vesicles/metabolism , Disease Models, Animal
4.
Int J Mol Sci ; 24(2)2023 Jan 14.
Article in English | MEDLINE | ID: mdl-36675188

ABSTRACT

Human bone marrow mesenchymal stem cell derived-extracellular vesicles (HBMSC-EV) are known for their regenerative and anti-inflammatory effects in animal models of myocardial ischemia. However, it is not known whether the efficacy of the EVs can be modulated by pre-conditioning of HBMSC by exposing them to either starvation or hypoxia prior to EV collection. HBMSC-EVs were isolated following normoxia starvation (NS), normoxia non-starvation (NNS), hypoxia starvation (HS), or hypoxia non-starvation (HNS) pre-conditioning. The HBMSC-EVs were characterized by nanoparticle tracking analysis, electron microscopy, Western blot, and proteomic analysis. Comparative proteomic profiling revealed that starvation pre-conditioning led to a smaller variety of proteins expressed, with the associated lesser effect of normoxia versus hypoxia pre-conditioning. In the absence of starvation, normoxia and hypoxia pre-conditioning led to disparate HBMSC-EV proteomic profiles. HNS HBMSC-EV was found to have the greatest variety of proteins overall, with 74 unique proteins, the greatest number of redox proteins, and pathway analysis suggestive of improved angiogenic properties. Future HBMSC-EV studies in the treatment of cardiovascular disease may achieve the most therapeutic benefits from hypoxia non-starved pre-conditioned HBMSC. This study was limited by the lack of functional and animal models of cardiovascular disease and transcriptomic studies.


Subject(s)
Cardiovascular Diseases , Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Humans , Cardiovascular Diseases/metabolism , Proteomics , Extracellular Vesicles/metabolism , Hypoxia/metabolism , Mesenchymal Stem Cells/metabolism
5.
Basic Res Cardiol ; 118(1): 3, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639609

ABSTRACT

Recent studies demonstrated that mitochondrial antioxidant MnSOD that reduces mitochondrial (mito) reactive oxygen species (ROS) helps maintain an optimal balance between sub-cellular ROS levels in coronary vascular endothelial cells (ECs). However, it is not known whether EC-specific mito-ROS modulation provides resilience to coronary ECs after a non-reperfused acute myocardial infarction (MI). This study examined whether a reduction in endothelium-specific mito-ROS improves the survival and proliferation of coronary ECs in vivo. We generated a novel conditional binary transgenic animal model that overexpresses (OE) mitochondrial antioxidant MnSOD in an EC-specific manner (MnSOD-OE). EC-specific MnSOD-OE was validated in heart sections and mouse heart ECs (MHECs). Mitosox and mito-roGFP assays demonstrated that MnSOD-OE resulted in a 50% reduction in mito-ROS in MHEC. Control and MnSOD-OE mice were subject to non-reperfusion MI surgery, echocardiography, and heart harvest. In post-MI hearts, MnSOD-OE promoted EC proliferation (by 2.4 ± 0.9 fold) and coronary angiogenesis (by 3.4 ± 0.9 fold), reduced myocardial infarct size (by 27%), and improved left ventricle ejection fraction (by 16%) and fractional shortening (by 20%). Interestingly, proteomic and Western blot analyses demonstrated upregulation in mitochondrial complex I and oxidative phosphorylation (OXPHOS) proteins in MnSOD-OE MHECs. These MHECs also showed increased mitochondrial oxygen consumption rate (OCR) and membrane potential. These findings suggest that mito-ROS reduction in EC improves coronary angiogenesis and cardiac function in non-reperfused MI, which are associated with increased activation of OXPHOS in EC-mitochondria. Activation of an energy-efficient mechanism in EC may be a novel mechanism to confer resilience to coronary EC during MI.


Subject(s)
Myocardial Infarction , Oxidative Phosphorylation , Mice , Animals , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , Endothelial Cells/metabolism , Proteomics , Myocardial Infarction/metabolism , Mitochondria/metabolism , Endothelium/metabolism
6.
Can J Physiol Pharmacol ; 101(2): 106-116, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36661235

ABSTRACT

Inflammatory pathways of Toll-like receptor 4 (TLR4) and NLRP3 inflammasome contribute to acute myocardial infarction (AMI) pathophysiology. The hypoxia-inducible factor 1α (HIF-1α), however, is a key transcription factor related to cardioprotection. This study aimed to compare the influence of carvedilol and thyroid hormones (TH) on inflammatory and HIF-1α proteins and on cardiac haemodynamics in the infarcted heart. Male Wistar rats were allocated into five groups: sham-operated group (SHAM), infarcted group (MI), infarcted treated with the carvedilol group (MI + C), infarcted treated with the TH group (MI + TH), and infarcted co-treated with the carvedilol and TH group (MI + C + TH). Haemodynamic analysis was assessed 15 days post-AMI. The left ventricle (LV) was collected for morphometric and Western blot analysis. The MI group presented LV systolic pressure reduction, LV end-diastolic pressure elevation, and contractility index decrease compared to the SHAM group. The MI + C, MI + TH, and MI + C + TH groups did not reveal such alterations compared to the SHAM group. The MI + TH and MI + C + TH groups presented reduced MyD88 and NLRP3 and increased HIF-1α levels. In conclusion, all treatments preserve the cardiac haemodynamic, and only TH, as isolated treatment or in co-treatment with carvedilol, was able to reduce MyD88 and NLRP3 and increase HIF-1α in the infarcted heart.


Subject(s)
Myeloid Differentiation Factor 88 , Myocardial Infarction , Animals , Male , Rats , Carvedilol/pharmacology , Carvedilol/therapeutic use , Myeloid Differentiation Factor 88/metabolism , Myocardial Infarction/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats, Wistar , Thyroid Hormones
7.
Vessel Plus ; 72023.
Article in English | MEDLINE | ID: mdl-38812773

ABSTRACT

Aim: To determine the differences in improvement in cardiac function by intramyocardial (IM) vs. intravenous (IV) injection of human bone mesenchymal stem cell-derived extracellular vesicles (HBMSC-EV) after acute MI. Methods: FVB mice underwent acute MI via left anterior descending coronary artery ligation and subsequent injection of: (1) IM saline control; (2) IM HBMSC-EV; (3) IV saline control; and (4) IV HBMSC-EV. Cardiac function was evaluated with weekly postoperative echocardiography. On postoperative day 28, the mice were euthanized, and the heart, lungs, liver, spleen, and kidneys were harvested. Given previous studies showing HBMSC-EV hepatic uptake after IV injection, the liver was evaluated for changes in inflammation, fibrosis, and proliferation. Results: On postoperative day 28, there were no significant differences in left ventricular ejection fraction (P = 0.6151), fractional shortening (P = 0.1135), or anterior border zone fibrosis (P = 0.6333) in any of the experimental groups. Interestingly, there was a strong trend demonstrating improvement in infarct size on fibrosis staining, which did not reach significance (P = 0.05620). There were no differences in hepatic inflammation, fibrosis, and proliferation. Conclusions: Although there was a trend in the improvement in infarct size, a single-dose administration of neither IM nor IV injection of HBMSC-EV resulted in significant improvement in post-MI cardiac function. A major limitation of this study is the lack of trials determining the optimal dose of HBMSC-EV needed in this model. However, the current study demonstrates that future studies are required to either optimize administration or bioengineer HBMSC-EV with cardiac-homing properties.

8.
An Acad Bras Cienc ; 93(suppl 4): e20210297, 2021.
Article in English | MEDLINE | ID: mdl-34706009

ABSTRACT

Acute myocardial infarction (AMI) is one of the major causes of heart failure and mortality. Glucocorticoids administration post-infarction has long been proposed, but it has shown conflicting results so far. This controversy may be associated with the glucocorticoid type and the period when it is administered. To elucidate these, the present aims to evaluate if the brief methylprednisolone acetate administration is determinant for heart adaptation after AMI. Male Wistar rats were divided into 3 groups: sham-operated (SHAM); infarcted (AMI); infarcted treated with methylprednisolone acetate (AMI+M). Immediately after surgery, the AMI+M group received a single dose of methylprednisolone acetate (40 mg/kg i.m.). After 56 days, the cardiac function was assessed and lungs, liver and heart were collected to determine rates of hypertrophy and congestion. Heart was used for oxidative stress and metalloproteinase activity analyses. Methylprednisolone acetate attenuated matrix metalloproteinase-2 activity, cardiac dilatation, and prevented the onset of pulmonary congestion, as well as avoided cardiac hypertrophy. Our data indicate that administration of methylprednisolone acetate shortly after AMI may be a therapeutic alternative for attenuation of detrimental ventricular remodeling.


Subject(s)
Methylprednisolone , Myocardial Infarction , Animals , Male , Matrix Metalloproteinase 2 , Methylprednisolone/therapeutic use , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardium , Rats , Rats, Wistar , Ventricular Remodeling
9.
Life Sci ; 284: 119917, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34478759

ABSTRACT

In addition to being an antioxidant, thioredoxin (Trx) is known to stimulate signaling pathways involved in cell proliferation and to inhibit apoptosis. The aim of this study was to explore the role of Trx in some of these pathways along the progression of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH). Male rats were first divided into two groups: monocrotaline (MCT - 60 mg/kg i.p.) and control (received saline), that were further divided into three groups: 1, 2, and 3 weeks. Animals were submitted to echocardiographic analysis. Right and left ventricles were used for the measurement of hypertrophy, through morphometric and histological analysis. The lung was prepared for biochemical and molecular analysis. One week after MCT injection, there was an increase in thioredoxin reductase (TrxR) activity, a reduction in glutathione reductase (GR) activity, and an increase in Trx-1 and vitamin D3 up-regulated protein-1 (VDUP-1) expression. Two weeks after MCT injection, there was an increase in VDUP-1, Akt and cleaved caspase-3 activation, and a decrease in Trx-1 and Nrf2 expression. PAH-induced by MCT promoted a reduction in Nrf2 and Trx-1 expression as well as an increase in Akt and VDUP-1 expression after three weeks. The increase in pulmonary vascular resistance was accompanied by increased TrxR activity, suggesting an association between the Trx system and functional changes in the progression of PAH. It seems that Trx-1 activation was an adaptive response to MCT administration to cope with pulmonary remodeling and disease progression, suggesting a potential new target for PAH therapeutics.


Subject(s)
Disease Progression , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Thioredoxins/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Cell Survival , Collagen/metabolism , Electrocardiography , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/pathology , Male , Monocrotaline , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Arterial Hypertension/complications , Pulmonary Arterial Hypertension/diagnostic imaging , Rats, Wistar
10.
STAR Protoc ; 2(3): 100753, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34458871

ABSTRACT

Reactive oxygen species (ROS) are implicated in endothelial dysfunction and cardiovascular disease. Endothelial cells (ECs) produce most ATP through glycolysis rather than oxidative phosphorylation; thus mitochondrial ROS production is lower than in other cell types. This makes quantification of changes in EC mitochondrial oxidative status challenging. Here, we present an optimized protocol using mitochondrial-targeted adenovirus-based redox sensor for ratiometric quantification of specific changes in mitochondrial ROS in live human coronary artery EC. For complete details on the use and execution of this protocol, please refer to Waypa et al. (2010); Liao et al. (2020); Gao et al. (2021).


Subject(s)
Coronary Vessels/cytology , Endothelial Cells/cytology , Green Fluorescent Proteins/genetics , Mitochondria/metabolism , Molecular Biology/methods , Adenoviridae/genetics , Cells, Cultured , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Green Fluorescent Proteins/metabolism , Humans , Mitochondria/genetics , Molecular Biology/instrumentation , Reactive Oxygen Species/metabolism , Transduction, Genetic
11.
Eur J Pharmacol ; 891: 173699, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33160936

ABSTRACT

The time-course of pulmonary arterial hypertension in the monocrotaline (MCT) model was investigated. Male rats were divided into two groups: MCT (received a 60 mg/kg i.p. injection) and control (received saline). The MCT and control groups were further divided into three cohorts, based on the follow-up interval: 1, 2, and 3 weeks. Right ventricle (RV) catheterization was performed and RV hypertrophy (RVH) was estimated. The lungs were used for biochemical, histological, molecular, and immunohistochemical analysis, while pulmonary artery rings were used for vascular reactivity. MCT promoted lung perivascular edema, inflammatory cells exudation, greater neutrophils and lymphocytes profile, and arteriolar wall thickness, compared to CTR group. Increases in pulmonary artery pressure and in RVH were observed in the MCT 2- and 3-week groups. The first week was marked by the presence of nitrosative stress (50% moderate and 33% accentuated staining by nitrotyrosine). These alterations lead to an adaptation of NO production by NO synthase activity after 2 weeks. Oxidative stress was evident in the third week, probably by an imbalance between endothelin-1 receptors, resulting in extracellular matrix remodeling, endothelial dysfunction, and RVH. Also, it was found a reduced pulmonary arterial vasodilatory response to acetylcholine after 2 (55%) and 3 (45%) weeks in MCT groups. The relevance of this study is precisely to show that nitrosative and oxidative stress predominate in distinct time windows of the disease progression.


Subject(s)
Lung/metabolism , Nitrosative Stress , Oxidative Stress , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Animals , Arterial Pressure , Disease Models, Animal , Disease Progression , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Lung/physiopathology , Male , Monocrotaline , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/physiopathology , Pulmonary Edema/etiology , Pulmonary Edema/metabolism , Pulmonary Edema/physiopathology , Rats, Wistar , Receptor, Endothelin A/metabolism , Time Factors , Vascular Remodeling , Vasodilation
12.
FEBS Open Bio ; 11(1): 35-47, 2021 01.
Article in English | MEDLINE | ID: mdl-33179452

ABSTRACT

Cardiovascular disease has been associated with increased levels of reactive oxygen species (ROS). Recently, we have shown that a critical balance between cytosolic ROS and mitochondrial ROS is crucial in cardiovascular health and that modulation of mitochondrial ROS helps prevent detrimental effects of cytosolic ROS on endothelial cells (EC) in transgenic animals. Here, we report the development of a controlled delivery system for a mitochondria-targeted antioxidant, JP4-039, from an electrospun scaffold made of FDA-approved biocompatible polymeric nanofibers. We demonstrate that the active antioxidant moiety was preserved in released JP4-039 for over 72 h using electron paramagnetic resonance. We also show that both the initial burst release of the drug within the first 20 min and the ensuing slow and sustained release that occurred over the next 24 h improved tube formation in human coronary artery ECs (HCAEC) in vitro. Taken together, these findings suggest that electrospinning methods can be used to upload mitochondrial antioxidant (JP4-039) onto a biocompatible nanofibrous PLGA scaffold, and the uploaded drug (JP4-039) retains nitroxide antioxidant properties upon release from the scaffold, which in turn can reduce mitochondrial ROS and improve EC function in vitro.


Subject(s)
Antioxidants/administration & dosage , Drug Carriers/chemistry , Nanofibers/chemistry , Nitrogen Oxides/administration & dosage , Antioxidants/pharmacokinetics , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cell Line , Coronary Vessels/cytology , Coronary Vessels/pathology , Drug Liberation , Endothelial Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Nitrogen Oxides/pharmacokinetics , Reactive Oxygen Species/metabolism
13.
J Cardiovasc Pharmacol ; 76(6): 698-707, 2020 12.
Article in English | MEDLINE | ID: mdl-33105324

ABSTRACT

Cellular death and survival signaling plays a key role in the progress of adverse cardiac remodeling after acute myocardial infarction (AMI). Therapeutic strategies, such as co-treatment with beta-blocker carvedilol and thyroid hormones (THs), give rise to new approaches that can sustain the cellular homeostasis after AMI. Therefore, we sought to investigate the effects of carvedilol and TH co-administration on apoptosis and survival proteins and on cardiac remodeling after AMI. Male Wistar rats were distributed in 5 groups as follows: sham-operated group (SHAM), infarcted group (MI), infarcted plus carvedilol group (MI+C), infarcted plus TH group (MI+TH), and infarcted plus carvedilol and TH co-treatment group (MI+C+TH). Echocardiographic analysis was performed, and hearts were collected for western blot evaluation. The MI group presented systolic posterior wall thickness loss, an increase in the wall tension index, and an increase in atrial natriuretic peptide tissue levels than the SHAM group. However, in the MI+C+TH group, these parameters were equally to the SHAM group. Moreover, whereas the MI group showed Bax protein expression elevated in relation to the SHAM group, the MI+C+TH group presented Bax reduction and also Akt activation compared with the MI group. In addition, the MI+TH group revealed beta-1 adrenergic receptor (ß1AR) upregulation compared with the MI and MI+C groups, whereas the MI+C+TH group presented lower levels of ß1AR in relation to the SHAM and MI+TH groups. In conclusion, we suggest that carvedilol and TH co-administration may mediate its cardioprotective effects against adverse cardiac remodeling post-AMI through the Bax reduction, Akt activation, and ß1AR decrease.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Carvedilol/pharmacology , Myocardial Infarction/drug therapy , Myocardium/metabolism , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Animals , Disease Models, Animal , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats, Wistar , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction
14.
Exp Physiol ; 105(9): 1561-1570, 2020 09.
Article in English | MEDLINE | ID: mdl-32667095

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does thyroid hormone treatment given after myocardial infarction preserve left ventricular function and treadmill exercise performance, and improve parameters of oxidative stress in the right ventricle and lungs of Wistar rats? What is the main finding and its importance? Thyroid hormone treatment improved the performance of the maximum exercise test in infarcted rats and induced effects in the heart and lungs that were similar to those observed with exercise training. This suggests there is a significant value of thyroid hormones for preserving exercise tolerance after myocardial infarction. ABSTRACT: Left ventricular myocardial infarction (MI) provokes damage in the heart and in other tissues, such as right ventricle and lungs. The present study elucidated whether thyroid hormone treatment (THT) may present positive effects in heart and lungs after MI, and whether or not these effects are similar to those of exercise training (ET). Male Wistar rats were divided into four groups: sham operated (SHAM), infarcted (MI), infarcted + exercise training (MIE), and infarcted + thyroid hormones (MIH). A maximum exercise test, left ventricle echocardiography, pulmonary histology, and oxidative stress in the right ventricle and lung were evaluated. THT and ET both reduced left ventricular dilatation and end-diastolic wall stress indexes to a similar extent. MI accentuated the content of macrophages and inflammatory infiltrate in the lungs, which was partially prevented in the MIH and MIE groups. THT and ET presented similar effects in the heart and lungs, and both improved the performance of the maximum exercise test in infarcted animals.


Subject(s)
Exercise Test , Myocardial Infarction/therapy , Physical Conditioning, Animal , Thyroid Hormones/pharmacology , Ventricular Function, Left , Animals , Echocardiography , Heart , Lung , Male , Myocardium , Oxidative Stress , Rats, Wistar
15.
Arch. endocrinol. metab. (Online) ; 63(3): 228-234, May-June 2019. tab, graf
Article in English | LILACS | ID: biblio-1011164

ABSTRACT

ABSTRACT Objective Hyperthyroidism causes many injuries in its target organs and the consequences are reflected systemically. As systemic alterations in hyperthyroidism at earlier stages have received partial attention, this study aimed to investigate systemic redox and inflammatory status at an early stage of T4-induced hyperthyroidism. Materials and methods Male Wistar rats were assigned to control and hyperthyroid groups (n = 7/group). The hyperthyroid group received L-thyroxine (12 mg/L) in their drinking water for 14 days whereas control group received only the vehicle. Body weight was measured on the 1st and 14th day of the protocol. On the 14th day, animals were anaesthetized. Blood was then collected from the retro-orbital venous plexus and then the animals were euthanised. The blood was separated into plasma and erythrocytes. Plasma was used to measure ROS levels, sulfhydryl compounds, IL-10, TNF-α and LDH levels; erythrocytes were used for the analysis of thioredoxin reductase activity, glutaredoxin content, and pentose cycle enzymes (total G6PD, G6PD and 6PGD). Results Hyperthyroid animals presented body weight gain and final body weight reduction, which was associated with increased ROS levels and decreased sulfhydryl content in plasma. Thioredoxin reductase activity, glutaredoxin content, and pentose cycle enzymes levels in erythrocytes, as well as IL-10, TNF-α and LDH plasma levels were unaltered. Conclusion Taken together, our results suggest an impairment in corporal mass associated with systemic oxidative stress at this stage of hyperthyroidism. Meanwhile, the pentose cycle was not influenced and systemic inflammation and tissue damage seem to be absent at this stage of hyperthyroidism.


Subject(s)
Animals , Male , Rats , Oxidative Stress/drug effects , Erythrocytes/metabolism , Hyperthyroidism/metabolism , Oxidation-Reduction , Pentoses , Thyroxine , Rats, Wistar , Disease Models, Animal , Erythrocytes/drug effects , Hyperthyroidism/blood , Antioxidants/metabolism
16.
J Clin Virol ; 116: 62-68, 2019 07.
Article in English | MEDLINE | ID: mdl-31103803

ABSTRACT

BACKGROUND: Acute respiratory infections caused by viruses are among the leading causes of morbidity and mortality. The inflammatory response that follows viral infection is important for the control of virus proliferation. However, if overwhelming, may be associated with complicated outcomes. OBJECTIVES: We assessed the clinical characteristics of patients with severe acute respiratory illness (SARI) evolving to acute respiratory distress syndrome (ARDS) and the factors related to death. STUDY DESIGN: Prospective study in 273 adult patients with SARI performed in a university-affiliated 800-bed hospital serving an area of epidemiologic vigilance of 102 municipalities and more than 2 million inhabitants. Influenza A (H1N1) 2009 (A/H1N1), influenza A H3N2, and influenza B were tested in all patients by RT-PCR. RESULTS: The overall hospital mortality rate was 17.6%. A total of 30.4% of patients tested positive for influenza A/H1N1. Patients with SARI that evolved to ARDS took significantly longer to take the first dose of oseltamivir (6.0 vs 1.0 days, p=0.002). Patients with H1N1 positive tests had almost 3 times higher probability of death, despite having significantly less comorbidities (p=0.027). The influenza A/H1N1 pdm09 vaccine reduced the odds of death by 78%. Nonsurvivors had a more intense inflammatory response than did survivors at 48 h (C-reactive protein: 31.0 ± 17.5 vs. 14.6 ± 8.9 mg/dl, p=0.001) as well as a more positive fluid balance. CONCLUSIONS: Hospital mortality associated with influenza H1N1-associated SARI and ARDS continued to be high years after the 2009 pandemic in a population with low vaccine coverage. Antiviral treatment started more than two days after onset of symptoms was more frequently associated with ARDS and death and, having had vaccine against influenza A (H1N1) was a factor independently related to survival.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/mortality , Influenza, Human/virology , Respiratory Tract Infections/mortality , Respiratory Tract Infections/virology , Adult , Aged , Antiviral Agents/therapeutic use , Female , Hospital Mortality , Humans , Inflammation/mortality , Inflammation/virology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza Vaccines/administration & dosage , Influenza, Human/drug therapy , Influenza, Human/pathology , Male , Middle Aged , Oseltamivir/therapeutic use , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/mortality , Respiratory Distress Syndrome/prevention & control , Respiratory Distress Syndrome/virology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/pathology , Risk Factors , Time-to-Treatment
17.
Arch Endocrinol Metab ; 63(3): 228-234, 2019.
Article in English | MEDLINE | ID: mdl-31038595

ABSTRACT

OBJECTIVE: Hyperthyroidism causes many injuries in its target organs and the consequences are reflected systemically. As systemic alterations in hyperthyroidism at earlier stages have received partial attention, this study aimed to investigate systemic redox and inflammatory status at an early stage of T4-induced hyperthyroidism. MATERIALS AND METHODS: Male Wistar rats were assigned to control and hyperthyroid groups (n = 7/group). The hyperthyroid group received L-thyroxine (12 mg/L) in their drinking water for 14 days whereas control group received only the vehicle. Body weight was measured on the 1st and 14th day of the protocol. On the 14th day, animals were anaesthetized. Blood was then collected from the retro-orbital venous plexus and then the animals were euthanised. The blood was separated into plasma and erythrocytes. Plasma was used to measure ROS levels, sulfhydryl compounds, IL-10, TNF-α and LDH levels; erythrocytes were used for the analysis of thioredoxin reductase activity, glutaredoxin content, and pentose cycle enzymes (total G6PD, G6PD and 6PGD). RESULTS: Hyperthyroid animals presented body weight gain and final body weight reduction, which was associated with increased ROS levels and decreased sulfhydryl content in plasma. Thioredoxin reductase activity, glutaredoxin content, and pentose cycle enzymes levels in erythrocytes, as well as IL-10, TNF-α and LDH plasma levels were unaltered. CONCLUSION: Taken together, our results suggest an impairment in corporal mass associated with systemic oxidative stress at this stage of hyperthyroidism. Meanwhile, the pentose cycle was not influenced and systemic inflammation and tissue damage seem to be absent at this stage of hyperthyroidism.


Subject(s)
Erythrocytes/metabolism , Hyperthyroidism/metabolism , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Disease Models, Animal , Erythrocytes/drug effects , Hyperthyroidism/blood , Male , Oxidation-Reduction , Pentoses , Rats , Rats, Wistar , Thyroxine
18.
Eur J Pharmacol ; 854: 159-166, 2019 Jul 05.
Article in English | MEDLINE | ID: mdl-30991047

ABSTRACT

After acute myocardial infarction (AMI), reactive oxygen species and oxidative stress have important roles in the progression to heart failure. As a therapeutic alternative, thyroid hormones (TH) revealed cardioprotective effects after AMI, including decreasing oxidative stress. Carvedilol beta-blocker, already used in the clinical treatment of AMI, also mitigate cardiac pathological remodelling. This study assessed the effects of post-AMI carvedilol and TH co-administration on oxidative stress and cardiac function as well as whether those effects were synergistic. Male Wistar rats were divided into five groups: sham-operated (SHAM), infarcted (MI), infarcted + TH (MI + TH), infarcted + carvedilol (MI + C) and infarcted + C + TH (MI + C + TH). Two days post-surgery, the SHAM and MI groups received saline, and treated groups received their respective treatments by gavage for 12 days. The animals were submitted to echocardiographic evaluation, ventricular catheterization and euthanized for heart collection to perform oxidative stress analysis. Treated groups improved for ejection fraction compared to the MI group. Carvedilol decreased the positive chronotropic TH effects in the MI + C + TH group. The MI and MI + C groups had increased reactive oxygen species and reduced sulfhydryl levels. Carvedilol and TH co-administration showed synergic effects in the MI + C + TH group, reducing reactive oxygen species levels and improving GSH/GSSG ratio. Moreover, co-treatment attenuated NADPH oxidase activity in the MI group. Therefore, this study showed for the first time that carvedilol and TH co-administration may improve redox balance and cardiac function after AMI. Such co-administration could represent a therapeutic strategy capable of preventing cardiac dysfunction and redox unbalance after AMI.


Subject(s)
Carvedilol/pharmacology , Heart/drug effects , Heart/physiopathology , Myocardial Infarction/metabolism , Oxidative Stress/drug effects , Thyroid Hormones/pharmacology , Animals , Antioxidants/metabolism , Drug Synergism , Electrocardiography/drug effects , Glutathione Disulfide/metabolism , Heart Rate/drug effects , Lipid Peroxidation/drug effects , Male , Myocardial Infarction/blood , Myocardial Infarction/physiopathology , NADPH Oxidases/metabolism , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Thyrotropin/blood
19.
J Biosci ; 43(5): 887-895, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30541949

ABSTRACT

Hyperthyroidism can lead to the activation of proteins which are associated with inflammation, apoptosis, hypertrophy, and heart failure. This study aimed to explore the inflammatory and apoptotic proteins involved in the hyperthyroidism-induced cardiac hypertrophy establishment. Male Wistar rats were divided into control and hyperthyroid (12 mg/L L-thyroxine, in drinking water for 28 days) groups. The expression of inflammatory and apoptotic signaling proteins was quantified in the left ventricle by Western blot. Hyperthyroidism was confirmed by evaluation of T3 and T4 levels, as well as cardiac hypertrophy development. There was no change in the expression of HSP70, HIF1-α, TNF-α, MyD88, p-NFκB, NFκB, p-p38, and p38. Reduced expression of p53 and PGC1-α was associated with increased TLR4 and decreased IL-10 expression. Decreased Bcl-2 expression and increased Bax/Bcl-2 ratio were also observed. The results suggest that reduced PGC1-α and IL-10, and elevated TLR4 proteins expression could be involved with the diminished mitochondrial biogenesis and anti-inflammatory response, as well as cell death signaling, in the establishment of hyperthyroidism-induced maladaptive cardiac hypertrophy.


Subject(s)
Cardiomegaly/genetics , Hyperthyroidism/genetics , Interleukin-10/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Toll-Like Receptor 4/genetics , Animals , Apoptosis/drug effects , Body Weight/drug effects , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Gene Expression Regulation , Heart/drug effects , Heart/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Hyperthyroidism/chemically induced , Hyperthyroidism/metabolism , Hyperthyroidism/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-10/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Organ Size/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Signal Transduction , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroxine/administration & dosage , Thyroxine/blood , Toll-Like Receptor 4/metabolism , Triiodothyronine/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
J Cardiovasc Pharmacol ; 72(5): 214-221, 2018 11.
Article in English | MEDLINE | ID: mdl-30212415

ABSTRACT

There is an increase in oxidative stress and apoptosis signaling during the transition from hypertrophy to right ventricular (RV) failure caused by pulmonary arterial hypertension (PAH) induced by monocrotaline (MCT). In this study, it was evaluated the action of copaiba oil on the modulation of proteins involved in RV apoptosis signaling in rats with PAH. Male Wistar rats (±170 g, n = 7/group) were divided into 4 groups: control, MCT, copaiba oil, and MCT + copaiba oil. PAH was induced by MCT (60 mg/kg intraperitoneally) and, 7 days later, treatment with copaiba oil (400 mg/kg by gavage) was given for 14 days. Echocardiographic and hemodynamic measurements were performed, and the RV was collected for morphometric evaluations, oxidative stress, apoptosis, and cell survival signaling, and eNOS protein expression. Copaiba oil reduced RV hypertrophy (24%), improved RV systolic function, and reduced RV end-diastolic pressure, increased total sulfhydryl levels and eNOS protein expression, reduced lipid and protein oxidation, and the expression of proteins involved in apoptosis signaling in the RV of MCT + copaiba oil as compared to MCT group. In conclusion, copaiba oil reduced oxidative stress, and apoptosis signaling in RV of rats with PAH, which may be associated with an improvement in cardiac function caused by this compound.


Subject(s)
Apoptosis/drug effects , Cardiovascular Agents/pharmacology , Fabaceae , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/prevention & control , Monocrotaline , Myocardium , Plant Oils/pharmacology , Ventricular Dysfunction, Right/prevention & control , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiovascular Agents/isolation & purification , Disease Models, Animal , Fabaceae/chemistry , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Plant Oils/isolation & purification , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Wistar , Signal Transduction/drug effects , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/pathology , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...