Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Physiol Rep ; 11(24): e15866, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38114067

RESUMO

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.


Assuntos
Diabetes Mellitus , Vesículas Extracelulares , Células-Tronco Mesenquimais , Animais , Humanos , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Células Cultivadas , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Diabetes Mellitus/metabolismo
2.
Can J Physiol Pharmacol ; 101(9): 447-454, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37581356

RESUMO

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.


Assuntos
Hipertensão Pulmonar , Óleos Voláteis , Hipertensão Arterial Pulmonar , Ratos , Masculino , Animais , Ratos Wistar , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Monocrotalina/efeitos adversos , Óxido Nítrico , Antioxidantes/farmacologia , Disponibilidade Biológica , Pulmão , Artéria Pulmonar , Hipertensão Pulmonar Primária Familiar , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/tratamento farmacológico , Óleos Voláteis/farmacologia , Modelos Animais de Doenças
3.
Physiol Rep ; 11(6): e15568, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36967241

RESUMO

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.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Infarto do Miocárdio , Camundongos , Animais , Humanos , Injeções Intravenosas , Distribuição Tecidual , Vesículas Extracelulares/metabolismo , Modelos Animais de Doenças
4.
Basic Res Cardiol ; 118(1): 3, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36639609

RESUMO

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.


Assuntos
Infarto do Miocárdio , Fosforilação Oxidativa , Camundongos , Animais , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/metabolismo , Células Endoteliais/metabolismo , Proteômica , Infarto do Miocárdio/metabolismo , Mitocôndrias/metabolismo , Endotélio/metabolismo
5.
Can J Physiol Pharmacol ; 101(2): 106-116, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36661235

RESUMO

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.


Assuntos
Fator 88 de Diferenciação Mieloide , Infarto do Miocárdio , Animais , Masculino , Ratos , Carvedilol/farmacologia , Carvedilol/uso terapêutico , Fator 88 de Diferenciação Mieloide/metabolismo , Infarto do Miocárdio/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ratos Wistar , Hormônios Tireóideos
6.
Int J Mol Sci ; 24(2)2023 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-36675188

RESUMO

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.


Assuntos
Doenças Cardiovasculares , Vesículas Extracelulares , Células-Tronco Mesenquimais , Animais , Humanos , Doenças Cardiovasculares/metabolismo , Proteômica , Vesículas Extracelulares/metabolismo , Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo
7.
An Acad Bras Cienc ; 93(suppl 4): e20210297, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34706009

RESUMO

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.


Assuntos
Metilprednisolona , Infarto do Miocárdio , Animais , Masculino , Metaloproteinase 2 da Matriz , Metilprednisolona/uso terapêutico , Infarto do Miocárdio/complicações , Infarto do Miocárdio/tratamento farmacológico , Miocárdio , Ratos , Ratos Wistar , Remodelação Ventricular
8.
Life Sci ; 284: 119917, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34478759

RESUMO

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.


Assuntos
Progressão da Doença , Hipertensão Arterial Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/patologia , Tiorredoxinas/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose , Sobrevivência Celular , Colágeno/metabolismo , Eletrocardiografia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Hipertrofia Ventricular Direita/complicações , Hipertrofia Ventricular Direita/patologia , Masculino , Monocrotalina , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Hipertensão Arterial Pulmonar/complicações , Hipertensão Arterial Pulmonar/diagnóstico por imagem , Ratos Wistar
9.
STAR Protoc ; 2(3): 100753, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34458871

RESUMO

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).


Assuntos
Vasos Coronários/citologia , Células Endoteliais/citologia , Proteínas de Fluorescência Verde/genética , Mitocôndrias/metabolismo , Biologia Molecular/métodos , Adenoviridae/genética , Células Cultivadas , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Mitocôndrias/genética , Biologia Molecular/instrumentação , Espécies Reativas de Oxigênio/metabolismo , Transdução Genética
10.
FEBS Open Bio ; 11(1): 35-47, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33179452

RESUMO

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.


Assuntos
Antioxidantes/administração & dosagem , Portadores de Fármacos/química , Nanofibras/química , Óxidos de Nitrogênio/administração & dosagem , Antioxidantes/farmacocinética , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/patologia , Linhagem Celular , Vasos Coronários/citologia , Vasos Coronários/patologia , Liberação Controlada de Fármacos , Células Endoteliais , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxidos de Nitrogênio/farmacocinética , Espécies Reativas de Oxigênio/metabolismo
11.
Eur J Pharmacol ; 891: 173699, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33160936

RESUMO

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.


Assuntos
Pulmão/metabolismo , Estresse Nitrosativo , Estresse Oxidativo , Hipertensão Arterial Pulmonar/metabolismo , Artéria Pulmonar/metabolismo , Animais , Pressão Arterial , Modelos Animais de Doenças , Progressão da Doença , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , Pulmão/fisiopatologia , Masculino , Monocrotalina , Hipertensão Arterial Pulmonar/induzido quimicamente , Hipertensão Arterial Pulmonar/fisiopatologia , Artéria Pulmonar/fisiopatologia , Edema Pulmonar/etiologia , Edema Pulmonar/metabolismo , Edema Pulmonar/fisiopatologia , Ratos Wistar , Receptor de Endotelina A/metabolismo , Fatores de Tempo , Remodelação Vascular , Vasodilatação
12.
J Cardiovasc Pharmacol ; 76(6): 698-707, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33105324

RESUMO

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.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Carvedilol/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Tiroxina/farmacologia , Tri-Iodotironina/farmacologia , Animais , Modelos Animais de Doenças , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Ratos Wistar , Receptores Adrenérgicos alfa 1/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Transdução de Sinais
13.
Exp Physiol ; 105(9): 1561-1570, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32667095

RESUMO

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.


Assuntos
Teste de Esforço , Infarto do Miocárdio/terapia , Condicionamento Físico Animal , Hormônios Tireóideos/farmacologia , Função Ventricular Esquerda , Animais , Ecocardiografia , Coração , Pulmão , Masculino , Miocárdio , Estresse Oxidativo , Ratos Wistar
14.
Arch. endocrinol. metab. (Online) ; 63(3): 228-234, May-June 2019. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1011164

RESUMO

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.


Assuntos
Animais , Masculino , Ratos , Estresse Oxidativo/efeitos dos fármacos , Eritrócitos/metabolismo , Hipertireoidismo/metabolismo , Oxirredução , Pentoses , Tiroxina , Ratos Wistar , Modelos Animais de Doenças , Eritrócitos/efeitos dos fármacos , Hipertireoidismo/sangue , Antioxidantes/metabolismo
15.
Arch Endocrinol Metab ; 63(3): 228-234, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31038595

RESUMO

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.


Assuntos
Eritrócitos/metabolismo , Hipertireoidismo/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Modelos Animais de Doenças , Eritrócitos/efeitos dos fármacos , Hipertireoidismo/sangue , Masculino , Oxirredução , Pentoses , Ratos , Ratos Wistar , Tiroxina
16.
J Clin Virol ; 116: 62-68, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31103803

RESUMO

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.


Assuntos
Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/mortalidade , Influenza Humana/virologia , Infecções Respiratórias/mortalidade , Infecções Respiratórias/virologia , Adulto , Idoso , Antivirais/uso terapêutico , Feminino , Mortalidade Hospitalar , Humanos , Inflamação/mortalidade , Inflamação/virologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vacinas contra Influenza/administração & dosagem , Influenza Humana/tratamento farmacológico , Influenza Humana/patologia , Masculino , Pessoa de Meia-Idade , Oseltamivir/uso terapêutico , Síndrome do Desconforto Respiratório/tratamento farmacológico , Síndrome do Desconforto Respiratório/mortalidade , Síndrome do Desconforto Respiratório/prevenção & controle , Síndrome do Desconforto Respiratório/virologia , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/patologia , Fatores de Risco , Tempo para o Tratamento
17.
Eur J Pharmacol ; 854: 159-166, 2019 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-30991047

RESUMO

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.


Assuntos
Carvedilol/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Infarto do Miocárdio/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Hormônios Tireóideos/farmacologia , Animais , Antioxidantes/metabolismo , Sinergismo Farmacológico , Eletrocardiografia/efeitos dos fármacos , Dissulfeto de Glutationa/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Infarto do Miocárdio/sangue , Infarto do Miocárdio/fisiopatologia , NADPH Oxidases/metabolismo , Oxirredução , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Compostos de Sulfidrila/metabolismo , Tireotropina/sangue
18.
J Biosci ; 43(5): 887-895, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30541949

RESUMO

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.


Assuntos
Cardiomegalia/genética , Hipertireoidismo/genética , Interleucina-10/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Receptor 4 Toll-Like/genética , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Regulação da Expressão Gênica , Coração/efeitos dos fármacos , Coração/fisiopatologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Hipertireoidismo/induzido quimicamente , Hipertireoidismo/metabolismo , Hipertireoidismo/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-10/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/patologia , Tiroxina/administração & dosagem , Tiroxina/sangue , Receptor 4 Toll-Like/metabolismo , Tri-Iodotironina/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
19.
J Cardiovasc Pharmacol ; 72(5): 214-221, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30212415

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Fabaceae , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Direita/prevenção & controle , Monocrotalina , Miocárdio , Óleos de Plantas/farmacologia , Disfunção Ventricular Direita/prevenção & controle , Função Ventricular Direita/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Fármacos Cardiovasculares/isolamento & purificação , Modelos Animais de Doenças , Fabaceae/química , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Óleos de Plantas/isolamento & purificação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Disfunção Ventricular Direita/induzido quimicamente , Disfunção Ventricular Direita/metabolismo , Disfunção Ventricular Direita/patologia , Proteína X Associada a bcl-2/metabolismo
20.
Free Radic Res ; 52(9): 988-999, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30203709

RESUMO

Oxidative stress alters signalling pathways for survival and cell death favouring the adverse remodelling of postmyocardial remnant cardiomyocytes, promoting functional impairment. The administration of pterostilbene (PTS), a phytophenol with antioxidant potential, can promote cardioprotection and represents a therapeutic alternative in acute myocardial infarction (AMI). The present study aims to explore the effects of oral administration of PTS complexed with hydroxypropyl-ß-cyclodextrin HPßCD (PTS:HPßCD complex) on the glutathione cycle, thiol protein activities and signalling pathways involving the protein kinase B (AKT) and glycogen synthase kinase-3ß (GSK-3ß) proteins in the left ventricle (LV) of infarcted rats. Animals were submitted to acute myocardial infarction through surgical ligation of the descending anterior branch of the left coronary artery and received over 8 days, by gavage, PTS:HPßCD complex at dose of 100 mg kg-1 day-1 (AMI + PTS group) or vehicle (aqueous solution with HPßCD) divided into Sham-operated (SHAM) and infarcted (AMI) groups. The results showed that the PBS: HPßCD complex decreased lipid peroxidation, prevented the decrease in thioredoxin reductase (TRxR) activity, and increased the activity of glutathione-S-transferase (GST) and glutaredoxin (GRx). Additionally, the expression of nuclear factor-erythroid two (Nrf2) and p-GSK-3ß was increased, whereas the p-GSK-3ß/GSK-3ß ratio was reduced in the LV of the infarcted animals. Overall, the PTS:HPßCD complex modulates activity of thiol-dependent enzymes and induces to the expression of antioxidant proteins, improving systolic function and mitigating the adverse cardiac remodelling post infarction.


Assuntos
Glicogênio Sintase Quinase 3 beta/genética , Infarto do Miocárdio/tratamento farmacológico , Fator 2 Relacionado a NF-E2/genética , Estilbenos/administração & dosagem , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Antioxidantes/administração & dosagem , Antioxidantes/química , Apoptose/efeitos dos fármacos , Cardiotônicos/administração & dosagem , Cardiotônicos/química , Ciclodextrinas/administração & dosagem , Ciclodextrinas/química , Modelos Animais de Doenças , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ratos , Estilbenos/química , Função Ventricular Esquerda/genética , Função Ventricular Esquerda/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA