Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Drug Deliv Transl Res ; 13(5): 1420-1435, 2023 05.
Article in English | MEDLINE | ID: mdl-36749480

ABSTRACT

The use of implantable biomaterials to replace physiological and anatomical functions has been widely investigated in the clinic. However, the selection of biomaterials is crucial for long-term function, and the implantation of certain biomaterials can cause inflammatory and fibrotic processes, triggering a foreign body reaction that leads to loss of function and consequent need for removal. Specifically, the Wnt signaling pathway controls the healing process of the human body, and its dysregulation can result in inflammation and fibrosis, such as in peritoneal fibrosis. Here, we assessed the effects of daily oral administration of a Wnt pathway inhibitor complex (CD:LGK974) to reduce the inflammatory, fibrotic, and angiogenic processes caused by intraperitoneal implants. CD:LGK974 significantly reduced the infiltration of immune cells and release of inflammatory cytokines in the implant region compared to the control groups. Furthermore, CD:LGK974 inhibited collagen deposition and reduced the expression of pro-fibrotic α-SMA and TGF-ß1, confirming fibrosis reduction. Finally, the CD:LGK974 complex decreased VEGF levels and both the number and area of blood vessels formed, suggesting decreased angiogenesis. This work introduces a potential new application of the Wnt inhibitor complex to reduce peritoneal fibrosis and the rejection of implants at the intraperitoneal site, possibly allowing for longer-term functionality of existing clinical biomaterials.


Subject(s)
Peritoneal Fibrosis , Humans , Peritoneal Fibrosis/complications , Vascular Endothelial Growth Factor A/metabolism , Inflammation/drug therapy , Inflammation/etiology , Foreign-Body Reaction/etiology , Foreign-Body Reaction/metabolism , Wound Healing
2.
Am J Physiol Cell Physiol ; 324(3): C606-C613, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36571443

ABSTRACT

The renin-angiotensin system (RAS) is a classical hormonal system involved in a myriad of cardiovascular functions. This system is composed of many different peptides that act in the heart through different receptors. One of the most important of these peptides is angiotensin II, which in pathological conditions triggers a set of actions that lead to heart failure. On the other hand, another RAS peptide, angiotensin-(1-7) is well known to develop powerful therapeutic effects in many forms of cardiac diseases. In the last decade, two new components of RAS were described, the heptapeptide alamandine and its receptor, the Mas-related G protein-coupled receptor member D (MrgD). Since then, great effort was made to characterize their physiological and pathological function in the heart. In this review, we summarize the latest insights about the actions of alamandine/MrgD axis in the heart, with particular emphasis in the cardiomyocyte. More specifically, we focused on their antihypertrophic and contractility effects, and the related molecular events activated in the cardiomyocyte.


Subject(s)
Myocytes, Cardiac , Receptors, G-Protein-Coupled , Myocytes, Cardiac/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Renin-Angiotensin System , Angiotensin II/metabolism , Angiotensin I/metabolism , Peptide Fragments/metabolism
3.
Peptides ; 158: 170862, 2022 12.
Article in English | MEDLINE | ID: mdl-35998722

ABSTRACT

Previous studies have suggested that the Angiotensin-(1-7) [(Ang-(1-7)] can change cardiac function by modulating the autonomic nervous system. However, it is unknown whether the Ang-(1-7) can modulate the effect of acetylcholine (ACh) in ventricular contractility. Thus, this study aimed to investigate whether Ang-(1-7) modifies the amplitude of the cardiac cholinergic effects and if these effects are intrinsic to the heart. In anesthetized Wistar rats, Ang-(1-7) attenuated the effect of ACh in decreasing the left ventricular end-systolic pressure (LVESP), dP/dtmax, and dP/dtmin, but did not modify the hypotensive effect of ACh. Similarly, Ang-(1-7) attenuated the reduction of the LVESP, dP/dtmax, and dP/dtmin evoked by ACh in isolated hearts. These effects were blocked by the Mas receptor antagonist, A-779, but not by the adenylyl cyclase inhibitor MDL-12,330 A. Ang-(1-7) also attenuated the reduction in the maximum contraction and relaxation speeds and the shortening promoted by ACh in isolated cardiomyocytes. These data show that Ang-(1-7) acting through Mas receptor counter-regulates the myocardial contractile response to ACh in an arterial pressure and heart rate-independent manner.


Subject(s)
Acetylcholine , Myocardial Contraction , Rats , Animals , Acetylcholine/pharmacology , Rats, Wistar , Heart , Myocytes, Cardiac , Angiotensin II/pharmacology
4.
Am J Physiol Heart Circ Physiol ; 320(1): H352-H363, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33124885

ABSTRACT

Alamandine is the newest identified peptide of the renin-angiotensin system (RAS) and has protective effects in the cardiovascular system. Although the involvement of classical RAS components in the genesis and progression of cardiac remodeling is well known, less is known about the effects of alamandine. Therefore, in the present study we investigated the effects of alamandine on cardiac remodeling induced by transverse aortic constriction (TAC) in mice. Male mice (C57BL/6), 10-12 wk of age, were divided into three groups: sham operated, TAC, and TAC + ALA (30 µg/kg/day alamandine for 14 days). The TAC surgery was performed under ketamine and xylazine anesthesia. At the end of treatment, the animals were submitted to echocardiographic examination and subsequently euthanized for tissue collection. TAC induced myocyte hypertrophy, collagen deposition, and the expression of matrix metalloproteinase (MMP)-2 and transforming growth factor (TGF)-ß in the left ventricle. These markers of cardiac remodeling were reduced by oral treatment with alamandine. Western blotting analysis showed that alamandine prevents the increase in ERK1/2 phosphorylation and reverts the decrease in 5'-adenosine monophosphate-activated protein kinase (AMPK)α phosphorylation induced by TAC. Although both TAC and TAC + ALA increased SERCA2 expression, the phosphorylation of phospholamban in the Thr17 residue was increased solely in the alamandine-treated group. The echocardiographic data showed that there are no functional or morphological alterations after 2 wk of TAC. Alamandine treatment prevents myocyte hypertrophy and cardiac fibrosis induced by TAC. Our results reinforce the cardioprotective role of alamandine and highlight its therapeutic potential for treating heart diseases related to pressure overload conditions.NEW & NOTEWORTHY Alamandine is the newest identified component of the renin-angiotensin system protective arm. Considering the beneficial effects already described so far, alamandine is a promising target for cardiovascular disease treatment. We demonstrated for the first time that alamandine improves many aspects of cardiac remodeling induced by pressure overload, including cell hypertrophy, fibrosis, and oxidative stress markers.


Subject(s)
Cardiovascular Agents/pharmacology , Heart Ventricles/drug effects , Hypertrophy, Left Ventricular/prevention & control , Oligopeptides/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Aorta/physiopathology , Aorta/surgery , Calcium-Binding Proteins/metabolism , Collagen/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Ligation , Male , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , Phosphorylation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
Biochem Pharmacol ; 182: 114210, 2020 12.
Article in English | MEDLINE | ID: mdl-32882205

ABSTRACT

TRPV1 is a cation channel expressed in peripheral nociceptive pathways and its activation can trigger nociception signals to the brain. Ketamine is an intravenous anesthetic routinely used for anesthesia induction and with potent analgesic activity. Despite its proven depressant action on peripheral sensory pathways, the relationship between ketamine and TRPV1 receptors is still unclear. In this study, we evaluated the effect of ketamine injected peripherally in a rat model of spontaneous pain induced by capsaicin. We also investigated the effect of ketamine on Ca2+ transients in cultured dorsal root ganglia (DRG) neurons and HEK293 cells expressing the TRPV1 receptor (HEK-TRPV1 cells). Intraplantar administration of ketamine caused an unexpected increase in nocifensive behavior induced by capsaicin. Incubation of HEK-TRPV1 cells with 10 µM ketamine increased TRPV1 and PKCє phosphorylation. Ketamine potentiated capsaicin-induced Ca2+ transients in HEK-TRPV1 cells and DRG neurons. Ketamine also prevented TRPV1 receptor desensitization induced by successive applications of capsaicin. єV1-2, a PKCє inhibitor, reduced potentiation of capsaicin-induced Ca2+ transients by ketamine. Taken together, our data indicate that ketamine potentiates TRPV1 receptor sensitivity to capsaicin through a mechanism dependent on PKCє activity.


Subject(s)
Ketamine/administration & dosage , Nociception/drug effects , Signal Transduction/drug effects , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism , Animals , Capsaicin/administration & dosage , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Amino Acid Antagonists/administration & dosage , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Male , Nociception/physiology , Rats , Rats, Wistar , Signal Transduction/physiology
7.
Proteomics Clin Appl ; 14(4): e2000017, 2020 07.
Article in English | MEDLINE | ID: mdl-32506788

ABSTRACT

PURPOSE: Although the pathophysiological response of cardiac tissue to pro-hypertrophic stimulus is well characterized, a comprehensive characterization of the molecular events underlying the pathological hypertrophy in cardiomyocytes during the early compensated cardiac hypertrophy is currently lacking. EXPERIMENTAL DESIGN: A quantitative label-free proteomic analysis of cardiomyocytes isolated was conducted from mice treated subcutaneously with isoproterenol (ISO) during 7 days in comparison with cardiomyocytes from control animals (CT). RESULTS: Canonical pathway analysis of dysregulated proteins indicated that ISO-hypertrophy drives the activation of actin cytoskeleton and integrin-linked kinase (ILK) signaling, and inhibition of the sirtuin signaling. Alteration in cardiac contractile function and calcium signaling are predicted as downstream effects of ISO-hypertrophy probably due to the upregulation of key elements such as myosin-7 (MYH7). Confocal microscopy corroborated that indeed ISO-treatment led to increased abundance of MYH7. Potential early markers for cardiac hypertrophy as APBB1, GOLGA4, HOOK1, KATNA1, KIFBP, MAN2B2, and SLC16A1 are also reported. CONCLUSIONS AND CLINICAL RELEVANCE: The data consist in a complete molecular mapping of ISO-induced compensated cardiac hypertrophy model at cardiomyocyte level. Marker candidates reported may assist early diagnosis of cardiac hypertrophy and ultimately heart failure.


Subject(s)
Cardiomegaly/metabolism , Isoproterenol/toxicity , Myocytes, Cardiac/metabolism , Proteome/genetics , Signal Transduction , Actins/genetics , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Myosin Heavy Chains/genetics , Protein Serine-Threonine Kinases/metabolism , Proteomics
8.
Am J Physiol Cell Physiol ; 318(4): C740-C750, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31913703

ABSTRACT

Overstimulation of the renin-angiotensin system (RAS) has been implicated in the pathogenesis of various cardiovascular diseases. Alamandine is a peptide newly identified as a protective component of the RAS; however, the mechanisms involved in its beneficial effects remain elusive. By using a well-characterized rat model of hypertension, the TGR (mREN2)27, we show that mREN ventricular myocytes are prone to contractile enhancement mediated by short-term alamandine (100 nmol/L) stimulation of Mas-related G protein-coupled receptor member D (MrgD) receptors, while Sprague-Dawley control cells showed no effect. Additionally, alamandine prevents the Ca2+ dysregulation classically exhibited by freshly isolated mREN myocytes. Accordingly, alamandine treatment of mREN myocytes attenuated Ca2+ spark rate and enhanced Ca2+ reuptake to the sarcoplasmic reticulum. Along with these findings, KN-93 fully inhibited the alamandine-induced increase in Ca2+ transient magnitude and phospholamban (PLN) phosphorylation at Thr17, indicating CaMKII as a downstream effector of the MrgD signaling pathway. In mREN ventricular myocytes, alamandine treatment induced significant nitric oxide (NO) production. Importantly, NO synthase inhibition prevented the contractile actions of alamandine, including PLN-Thr17 phosphorylation at the CaMKII site, thereby indicating that NO acts upstream of CaMKII in the alamandine downstream signaling. Altogether, our results show that enhanced contractile responses mediated by alamandine in cardiomyocytes from hypertensive rats occur through a NO-dependent activation of CaMKII.


Subject(s)
Myocytes, Cardiac/drug effects , Nitric Oxide/metabolism , Oligopeptides/pharmacology , Sarcoplasmic Reticulum/drug effects , Animals , Calcium-Binding Proteins , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Male , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Rats , Sarcoplasmic Reticulum/metabolism , Signal Transduction/drug effects
10.
Brain Behav Immun ; 81: 444-454, 2019 10.
Article in English | MEDLINE | ID: mdl-31271871

ABSTRACT

The gastrointestinal (GI) tract harbors commensal microorganisms as well as invasive bacteria, toxins and other pathogens and, therefore, plays a pivotal barrier and immunological role against pathogenic agents. The vagus nerve is an important regulator of the GI tract-associated immune system, having profound effects on inflammatory responses. Among GI tract organs, the liver is a key site of immune surveillance, as it has a large population of resident macrophages and receives the blood drained from the guts through the hepatic portal circulation. Although it is widely accepted that the hepatic tissue is a major target for vagus nerve fibers, the role of this neural circuit in liver immune functions is still poorly understood. Herein we used in vivo imaging techniques, including confocal microscopy and scintigraphy, to show that vagus nerve stimulation increases the phagocytosis activity by resident macrophages in the liver, even on the absence of an immune challenge. The activation of this neural circuit in a non-lethal model of sepsis optimized the removal of bacteria in the liver and resulted in the production of anti-inflammatory and pro-regenerative cytokines. Our findings provide new insights into the neural regulation of the immune system in the liver.


Subject(s)
Liver/immunology , Phagocytosis/physiology , Vagus Nerve/physiology , Animals , Cytokines , Female , Gastrointestinal Tract , Liver/pathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phagocytes/metabolism , Sepsis/immunology , Vagus Nerve/pathology , Vagus Nerve Stimulation/methods
11.
Eur J Pharmacol ; 859: 172488, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31233746

ABSTRACT

Cardiac arrhythmias are among the most important pathologies that lead to sudden death. The discovery of new therapeutic options against arrhythmias with low adverse effects is of paramount importance. Farnesol is found in essential oils with antioxidant, anti-inflammatory and cardioprotective properties. The aim of this work was to investigate the effects of farnesol on the contractile and electrophysiological properties in rat heart and evaluate its antiarrhythmic action. It was evaluated farnesol effects on the left ventricular developed pressure, ECG, potassium (Ik) and L-type Ca2+ currents (ICa,L), action potential, intracellular Ca2+ transient, Ca2+ sparks and waves and reactive oxygen species production. Antiarrhythmic activity of farnesol was determined in vivo and ex vivo. The results showed that 50 µM farnesol did not alter left ventricular developed pressure, heart rate, ECG parameters and intracellular Ca2+ transient but reduced ICa,L. Farnesol reduced action potential duration at 90% repolarization. Notably, farnesol improved arrhythmia score and the incidence of the most severe arrhythmias. Farnesol attenuated the generation of reactive oxygen species, Ca2+ sparks and waves in isolated cardiomyocytes submitted to Ca2+ overload. In conclusion, farnesol has antiarrhythmic effect mediated by reducing of ICa,L and IK along with a decrease of reactive oxygen species production and normalized Ca2+ sparks and waves.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Calcium Channel Blockers/pharmacology , Calcium/metabolism , Farnesol/pharmacology , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium Channel Blockers/therapeutic use , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Electrocardiography/drug effects , Farnesol/therapeutic use , Intracellular Space/drug effects , Intracellular Space/metabolism , Male , Myocardial Contraction/drug effects , Oxygen/metabolism , Potassium/metabolism , Rats , Rats, Wistar , Ventricular Dysfunction, Left/drug therapy
12.
Life Sci ; 228: 121-127, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31039364

ABSTRACT

AIMS: B1- and B2-kinin receptors play a major role in several cardiovascular diseases. Therefore, we aimed to evaluate cardiac functional consequences of B1- and B2-kinin receptors ablation, focusing on the cardiac ROS and NO generation. MAIN METHODS: Cardiac contractility, ROS, and NO generation, and protein expression were evaluated in male wild-type (WT), B1- (B1-/-) and B2-kinin (B2-/-) knockout mice. KEY FINDINGS: Impaired contractility in B1-/- and B2-/- hearts was associated with oxidative stress through upregulation of NADPH oxidase p22phox subunit. B1-/- and B2-/- hearts presented higher NO and peroxynitrite levels than WT. Despite decreased sarcoplasmic reticulum Ca2+ ATPase pump (SERCA2) expression, nitration at tyrosine residues of SERCA2 was markedly higher in B1-/- and B2-/- hearts. SIGNIFICANCE: B1- and B2-kinin receptors govern ROS generation, while disruption of B1- and B2-kinin receptors leads to impaired cardiac dysfunction through excessive tyrosine nitration on the SERCA2 structure.


Subject(s)
Heart Diseases/genetics , Heart/physiopathology , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/genetics , Animals , Gene Deletion , Heart Diseases/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism
13.
Free Radic Biol Med ; 130: 174-188, 2019 01.
Article in English | MEDLINE | ID: mdl-30315935

ABSTRACT

We previously demonstrated that the loss of female hormones induces cardiac and mitochondrial dysfunction in the female heart. Here, we show the impact of endurance training for twelve weeks, a nonpharmacological therapy against cardiovascular disease caused by ovariectomy and its contribution to cardiac contractility, mitochondrial quality control, bioenergetics and oxidative damage. We found that ovariectomy induced cardiac hypertrophy and dysfunction by decreasing SERCA2 and increasing phospholamban protein expression. Endurance training restored myocardial contractility, SERCA2 levels, increased calcium transient in ovariectomized rats but did not change phospholamban protein expression or cardiac hypertrophy. Additionally, ovariectomy decreased the amount of intermyofibrillar mitochondria and induced mitochondrial fragmentation that were accompanied by decreased levels of mitofusin 1, PGC-1α, NRF-1, total AMPK-α and mitochondrial Tfam. Endurance training prevented all these features except for mitofusin 1. Ovariectomy reduced O2 consumption, elevated O2.- release and increased Ca2+-induced mitochondrial permeability transition pore opening in both mitochondrial subpopulations. Ovariectomy also increased NOX-4 protein expression in the heart, reduced mitochondrial Mn-SOD, catalase protein expression and increased protein carbonylation in both mitochondrial subpopulations, which were prevented by endurance training. Taken together, our findings show that endurance training prevented cardiac contractile dysfunction and mitochondrial quality control in ovariectomized rats.


Subject(s)
Cardiomegaly/prevention & control , Endurance Training , Mitochondria, Heart/metabolism , Myocardium/metabolism , Physical Conditioning, Animal , Animals , Cardiomegaly/etiology , Cells, Cultured , Energy Metabolism , Female , Gonadal Steroid Hormones/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Contraction , Ovariectomy/adverse effects , Oxidative Stress , Rats , Rats, Wistar , Recovery of Function , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
14.
FEBS J ; 286(1): 110-123, 2019 01.
Article in English | MEDLINE | ID: mdl-30451379

ABSTRACT

Huntington's disease (HD) is a neurodegenerative genetic disorder. Although described as a brain pathology, there is evidence suggesting that defects in other systems can contribute to disease progression. In line with this, cardiovascular defects are a major cause of death in HD. To date, relatively little is known about the peripheral abnormalities associated with the disease. Here, we applied a range of assays to evaluate cardiac electro-mechanical properties in vivo, using a previously characterized mouse model of HD (BACHD), and in vitro, using cardiomyocytes isolated from the same mice. We observed conduction disturbances including QT interval prolongation in BACHD mice, indicative of cardiac dysfunction. Cardiomyocytes from these mice demonstrated cellular electro-mechanical abnormalities, including a prolonged action potential, arrhythmic contractions, and relaxation disturbances. Cellular arrhythmia was accompanied by an increase in calcium waves and increased Ca2+ /calmodulin-dependent protein kinase II activity, suggesting that disruption of calcium homeostasis plays a key part. We also described structural abnormalities in the mitochondria of BACHD-derived cardiomyocytes, indicative of oxidative stress. Consistent with this, imbalances in superoxide dismutase and glutathione peroxidase activities were detected. Our data provide an in vivo demonstration of cardiac abnormalities in HD together with new insights into the cellular mechanistic basis, providing a possible explanation for the higher cardiovascular risk in HD.


Subject(s)
Arrhythmias, Cardiac/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Huntington Disease/physiopathology , Mitochondria/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Animals , Antioxidants/metabolism , Arrhythmias, Cardiac/metabolism , Biomechanical Phenomena , Electrophysiological Phenomena , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Phosphorylation
15.
J Mol Cell Cardiol ; 125: 61-72, 2018 12.
Article in English | MEDLINE | ID: mdl-30339842

ABSTRACT

BACKGROUND: Currently viewed as a complementary non-pharmacological intervention for preventing cardiac disorders, long-term aerobic training produces cardioprotection through remote ischemic preconditioning (RIPC) mechanisms. However, RIPC triggered by acute exercise remains poorly understood. Although resistance exercise (RE) has been highly recommended by several public health guidelines, there is no evidence showing that RE mediates RIPC. Hence, we investigated whether RE induces cardiac RIPC through nitric oxide synthase (NOS)-dependent mechanism. METHODS AND RESULTS: Acute RE at 40% of the maximal load augmented systemic nitrite levels, associated with increased cardiac eNOS phosphorylation, without affecting nNOS activity. Using an experimental model of myocardial infarction (MI) through ischemia-reperfusion (IR), RE fully prevented the loss of cardiac contractility and the extent of MI size compared to non-exercised (NE) rats. Moreover, RE mitigated aberrant ST-segment and reduced life-threatening arrhythmias induced by IR. Importantly, inhibition of NOS abolished the RE-mediated cardioprotection. After IR, NE rats showed increased cardiac eNOS activity, associated with reduced dimer/monomer ratio. Supporting the pivotal role of eNOS coupling during MI, non-exercised rats displayed a marked generation of reactive oxygen species (ROS) and oxidative-induced carbonylation of proteins, whereas RE prevented these responses. We validated our data demonstrating a restoration of physiological ROS levels in NE + IR cardiac sections treated with BH4, a cofactor oxidatively depleted during eNOS uncoupling, while cardiac ROS generation from exercised rats remained unchanged, suggesting no physiological needs of supplemental eNOS cofactors. CONCLUSION: Together, our findings strongly indicate that RE mediates RIPC by limiting eNOS uncoupling and mitigates myocardial IR injury.


Subject(s)
Ischemic Preconditioning/methods , Nitric Oxide Synthase Type III/metabolism , Physical Conditioning, Animal/physiology , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Blotting, Western , Electrocardiography , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
16.
Am J Physiol Cell Physiol ; 314(6): C702-C711, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29443552

ABSTRACT

The renin-angiotensin system (RAS) plays a pivotal role in the pathogenesis of cardiovascular diseases. New members of this system have been characterized and shown to have biologically relevant actions. Alamandine and its receptor MrgD are recently identified components of RAS. In the cardiovascular system, alamandine actions included vasodilation, antihypertensive, and antifibrosis effects. Currently, the actions of alamandine on cardiomyocytes are unknown. Here our goal was twofold: 1) to unravel the signaling molecules activated by the alamandine/MrgD axis in cardiomyocytes; and 2) to evaluate the ability of this axis to prevent angiotensin II (ANG II)-induced hypertrophy. In cardiomyocytes from C57BL/6 mice, alamandine treatment induced an increase in nitric oxide (NO) production, which was blocked by d-Pro7-ANG-(1-7), a MrgD antagonist. This NO rise correlated with increased phosphorylation of AMPK. Alamandine-induced NO production was preserved in Mas-/- myocytes and lost in MrgD-/- cells. Binding of fluorescent-labeled alamandine was observed in wild-type cells, but it was dramatically reduced in MrgD-/- myocytes. We also assessed the consequences of prolonged alamandine exposure to cultured neonatal rat cardiomyocytes (NRCMs) treated with ANG II. Treatment of NRCMs with alamandine prevented ANG II-induced hypertrophy. Moreover, the antihypertrophic actions of alamandine were mediated via MrgD and NO, since they could be prevented by d-Pro7-ANG-(1-7) or inhibitors of NO synthase or AMPK. ß-Alanine, a MrgD agonist, recapitulated alamandine's cardioprotective effects in cardiomyocytes. Our data show that alamandine via MrgD induces AMPK/NO signaling to counterregulate ANG II-induced hypertrophy. These findings highlight the therapeutic potential of the alamandine/MrgD axis in the heart.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Angiotensin II/toxicity , Cardiomegaly/prevention & control , Myocytes, Cardiac/drug effects , Nerve Tissue Proteins/agonists , Nitric Oxide/metabolism , Oligopeptides/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cells, Cultured , Enzyme Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/metabolism , Oligopeptides/metabolism , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
17.
Food Chem Toxicol ; 111: 557-566, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29208507

ABSTRACT

Myrtenol is a monoterpene with multiple pharmacological activities. However, although monoterpenes have been proposed to play beneficial roles in a variety of cardiac disorders, pharmacological actions of myrtenol in the heart are not yet reported. Hence, the aim of this study was to evaluate whether myrtenol promotes cardioprotection against myocardial ischemia-reperfusion (IR) injury, and the mechanisms involved in these effects. Male Wistar rats were orally treated for seven consecutive days with myrtenol (50 mg/kg) or N-acetyl cysteine (1.200 mg/kg, NAC). Afterward, hearts were subjected to myocardial IR injury. Here, we show that the severe impairment of contractile performance induced by IR was significantly prevented by myrtenol or NAC. Moreover, myrtenol abolished aberrant electrocardiographic waveform (ST-segment elevation), as well as reduced life-threatening arrhythmias and infarct size induced by IR injury. Importantly, myrtenol fully prevented the massive increase of cardiac reactive oxygen species generation and oxidative stress damage. Accordingly, myrtenol restored the impairment of endogenous antioxidant enzymes (superoxide dismutase, catalase, glutathione peroxidase and reductase) activities and balance of pro- and anti-apoptotic pathways (Bax and Bcl-2), associated with decreased apoptotic cells. Taken together, our data show that myrtenol promotes cardioprotection against IR injury through attenuation of oxidative stress and inhibition of pro-apoptotic pathway.


Subject(s)
Antioxidants/metabolism , Apoptosis/drug effects , Monoterpenes/administration & dosage , Myocardial Reperfusion Injury/prevention & control , Animals , Bicyclic Monoterpenes , Catalase/metabolism , Glutathione Peroxidase/metabolism , Heart/drug effects , Heart/physiopathology , Humans , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
18.
Fisioter. Mov. (Online) ; 30(3): 443-451, July-Sept. 2017. tab
Article in English | LILACS | ID: biblio-891993

ABSTRACT

Abstract Introduction: Osteoarthritis is a disease that affects millions of Brazilians.Therapeutic ultrasound has been used in its treatment, either alone or associated with drugs. Objective: The aim of this study was to evaluate the effects of ultrasound (US) associated with Copaiba oil (CO) on knee osteoarthritis. Methods: Patients were divided into three different groups: US, US+CO, CO.Ten treatment sessions were held twice a week, 30 minutes each.Pain intensity was assessed through the Visual Analog Scale (VAS) and Range of Motion (ROM) by goniometry, and muscle strength was assessed by means of the Medical Research Council Scale. Statistical analysis was performed by Cohen's d test, student's t test and ANOVA, considering p<0.05 as significant. Results: Pain reduced in all groups.The US+CO group (d = -3.50) presented larger effect size when compared to the other groups. Regarding ROM, the largest effect size was observed in the US+CO group for flexion (d = 0.86) and extension (d = 0.97) in comparison with the remainder groups. Muscle strength increased in the US (d = 1.54) and US+CO (d = 1.60) groups for flexion.Regarding extension, the US group presented the largest effect size (d = 1.80). Conclusion: Therapeutic ultrasound associated with copaiba oil is a practical and effective therapy for the treatment of inflammatory diseases such as osteoarthritis.


Resumo Introdução: A osteoartrite é uma doença que afeta milhões de brasileiros. O ultrassom terapêutico tem sido utilizado em seu tratamento tanto sozinho, quanto associado a fármacos. Objetivo: Avaliar o efeito do ultrassom (US) associado ao óleo de copaíba (OC) em pacientes com osteoartrite de joelho. Métodos: Os pacientes foram distribuídos em 3 grupos distintos: US, US+OC e OC. Foram realizadas 10 sessões de tratamento, duas vezes por semana durante 30 minutos. A intensidade da dor foi avaliada pela Escala Visual Analógica da Dor (EVA), amplitude de movimento ADM pela goniometria e força muscular pelo Score Medical Research Council. A análise estatística foi feita pelo Teste T de Student e ANOVA e a magnitude do efeito (d), considerando p<0,05 como valores significativos. Resultados: A dor foi atenuada em todos os grupos, sendo apresentada uma magnitude maior do efeito para o grupo US+OC (d = -3,50) quando comparado aos demais grupos. Em relação a ADM a magnitude do efeito foi maior no grupo US+OC (d= 0,86) para a flexão e extensão (d = 0,97) quando comparados com os outros grupos. Na variável força muscular os grupos US (d= 1,54) e US+OC (d = 1,60) foram mais eficazes no movimento de flexão e na extensão, o grupo US exibiu o maior tamanho de efeito (d = 1,80) quando comparados aos demais grupos. Conclusão: O ultrassom terapêutico associado ao óleo de copaíba é uma terapia efetiva e prática para o tratamento de doenças inflamatórias, tais como a osteoartrite.

19.
Basic Clin Pharmacol Toxicol ; 120(6): 550-559, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27992670

ABSTRACT

Quercetin is a plant flavonoid with several biological activities. This study aimed to describe quercetin effects on contractile and electrophysiological properties of the cardiac muscle as well as on calcium handling. Quercetin elicited positive inotropism that was significantly reduced by propranolol indicating an involvement of the sympathetic nervous system. In cardiomyocytes, 30 µM quercetin increased ICa,L at 0 mV from -0.95 ± 0.01 A/F to -1.21 ± 0.08 A/F. The membrane potential at which 50% of the channels are activated (V0.5 ) shifted towards more negative potentials from -13.06 ± 1.52 mV to -19.26 ± 1.72 mV and did not alter the slope factor. Furthermore, quercetin increased [Ca2+ ]i transient by 28% when compared to control. Quercetin accelerated [Ca2+ ]i transient decay time, which could be attributed to SERCA activation. In resting cardiomyocytes, quercetin did not change amplitude or frequency of Ca2+ sparks. In isolated heart, quercetin increased heart rate and decreased PRi, QTc and duration of the QRS complex. Thus, we showed that quercetin activates ß-adrenoceptors, leading to increased L-type Ca2+ current and cell-wide intracellular Ca2+ transient without visible changes in Ca2+ sparks.


Subject(s)
Heart/drug effects , Quercetin/pharmacology , Animals , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/physiology , Electrocardiography/drug effects , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocardium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...