Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Physiol ; 9: 521, 2018.
Article in English | MEDLINE | ID: mdl-29867564

ABSTRACT

G protein-coupled estrogen receptor (GPER) is an estrogen receptor expressed in the cardiovascular system. G1, a selective GPER ligand, exerts cardiovascular effects through activation of the PI3K-Akt pathway and Notch signaling in normotensive animals. Here, we investigated whether the G1/GPER interaction is involved in the limitation of infarct size, and improvement of post-ischemic contractile function in female spontaneous hypertensive rat (SHR) hearts. In this model, we also studied Notch signaling and key components of survival pathway, namely PI3K-Akt, nitric oxide synthase (NOS) and mitochondrial K+-ATP (MitoKATP) channels. Rat hearts isolated from female SHR underwent 30 min of global, normothermic ischemia and 120 min of reperfusion. G1 (10 nM) alone or specific inhibitors of GPER, PI3K/NOS and MitoKATP channels co-infused with G1, just before I/R, were studied. The involvement of Notch1 was studied by Western blotting. Infarct size and left ventricular pressure were measured. To confirm endothelial-independent G1-induced protection by Notch signaling, H9c2 cells were studied with specific inhibitor, N-[N-(3,5 difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT, 5 µM), of this signaling. Using DAPT, we confirmed the involvement of G1/Notch signaling in limiting infarct size in heart of normotensive animals. In the hypertensive model, G1-induced reduction in infarct size and improvement of cardiac function were prevented by the inhibition of GPER, PI3K/NOS, and MitoKATP channels. The involvement of Notch was confirmed by western blot in the hypertensive model and by the specific inhibitor in the normotensive model and cardiac cell line. Our results suggest that GPERs play a pivotal role in mediating preconditioning cardioprotection in normotensive and hypertensive conditions. The G1-induced protection involves Notch1 and is able to activate the survival pathway in the presence of comorbidity. Several pathological conditions, including hypertension, reduce the efficacy of ischemic conditioning strategies. However, G1-induced protection can result in significant reduction of I/R injury also female in hypertensive animals. Further studies may ascertain the clinical translation of the present results.

2.
Front Physiol ; 9: 366, 2018.
Article in English | MEDLINE | ID: mdl-29706897

ABSTRACT

Changes in environmental oxygen (O2) are naturally occurring phenomena which ectotherms have to face on. Many species exhibit a striking capacity to survive and remain active for long periods under hypoxia, even tolerating anoxia. Some fundamental adaptations contribute to this capacity: metabolic suppression, tolerance of pH and ionic unbalance, avoidance and/or repair of free-radical-induced cell injury during reoxygenation. A remarkable feature of these species is their ability to preserve a normal cardiovascular performance during hypoxia/anoxia to match peripheral (tissue pO2) requirements. In this review, we will refer to paradigms of hypoxia- and anoxia-tolerant teleost fish to illustrate cardiac physiological strategies that, by involving nitric oxide and its metabolites, play a critical role in the adaptive responses to O2 limitation. The information here reported may contribute to clarify the molecular and cellular mechanisms underlying heart vulnerability vs. resistance in relation to O2 availability.

3.
J Cell Mol Med ; 21(12): 3670-3678, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28744974

ABSTRACT

Patients with ischaemic heart disease or chronic heart failure show altered levels of obestatin, suggesting a role for this peptide in human heart function. We have previously demonstrated that GH secretagogues and the ghrelin gene-derived peptides, including obestatin, exert cardiovascular effects by modulating cardiac inotropism and vascular tone, and reducing cell death and contractile dysfunction in hearts subjected to ischaemia/reperfusion (I/R), through the Akt/nitric oxide (NO) pathway. However, the mechanisms underlying the cardiac actions of obestatin remain largely unknown. Thus, we suggested that obestatin-induced activation of PI3K/Akt/NO and PKG signalling is implicated in protection of the myocardium when challenged by adrenergic, endothelinergic or I/R stress. We show that obestatin exerts an inhibitory tone on the performance of rat papillary muscle in both basal conditions and under ß-adrenergic overstimulation, through endothelial-dependent NO/cGMP/PKG signalling. This pathway was also involved in the vasodilator effect of the peptide, used both alone and under stress induced by endothelin-1. Moreover, when infused during early reperfusion, obestatin reduced infarct size in isolated I/R rat hearts, through an NO/PKG pathway, comprising ROS/PKC signalling, and converging on mitochondrial ATP-sensitive potassium [mitoK(ATP)] channels. Overall, our results suggest that obestatin regulates cardiovascular function in stress conditions and induces cardioprotection by mechanisms dependent on activation of an NO/soluble guanylate cyclase (sGC)/PKG pathway. In fact, obestatin counteracts exaggerated ß-adrenergic and endothelin-1 activity, relevant factors in heart failure, suggesting multiple positive effects of the peptide, including the lowering of cardiac afterload, thus representing a potential candidate in pharmacological post-conditioning.


Subject(s)
Cardiotonic Agents/pharmacology , Myocardial Infarction/prevention & control , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide/metabolism , Peptide Hormones/pharmacology , Animals , Cardiotonic Agents/chemistry , Cardiotonic Agents/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Endothelin-1/antagonists & inhibitors , Endothelin-1/pharmacology , Gene Expression Regulation , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Male , Myocardial Contraction/drug effects , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Organ Culture Techniques , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Papillary Muscles/pathology , Peptide Hormones/genetics , Peptide Hormones/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction , Soluble Guanylyl Cyclase/genetics , Soluble Guanylyl Cyclase/metabolism
4.
Prog Neurobiol ; 154: 37-61, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28442394

ABSTRACT

The granin family comprises altogether 7 different proteins originating from the diffuse neuroendocrine system and elements of the central and peripheral nervous systems. The family is dominated by three uniquely acidic members, namely chromogranin A (CgA), chromogranin B (CgB) and secretogranin II (SgII). Since the late 1980s it has become evident that these proteins are proteolytically processed, intragranularly and/or extracellularly into a range of biologically active peptides; a number of them with regulatory properties of physiological and/or pathophysiological significance. The aim of this comprehensive overview is to provide an up-to-date insight into the distribution and properties of the well established granin-derived peptides and their putative roles in homeostatic regulations. Hence, focus is directed to peptides derived from the three main granins, e.g. to the chromogranin A derived vasostatins, betagranins, pancreastatin and catestatins, the chromogranin B-derived secretolytin and the secretogranin II-derived secretoneurin (SN). In addition, the distribution and properties of the chromogranin A-derived peptides prochromacin, chromofungin, WE14, parastatin, GE-25 and serpinins, the CgB-peptide PE-11 and the SgII-peptides EM66 and manserin will also be commented on. Finally, the opposing effects of the CgA-derived vasostatin-I and catestatin and the SgII-derived peptide SN on the integrity of the vasculature, myocardial contractility, angiogenesis in wound healing, inflammatory conditions and tumors will be discussed.


Subject(s)
Chromogranins/metabolism , Animals , Humans , Peptides/metabolism
5.
J Cell Physiol ; 232(7): 1640-1649, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27607345

ABSTRACT

The use of Doxorubicin (Dox), a frontline drug for many cancers, is often complicated by dose-limiting cardiotoxicity in approximately 20% of patients. The G-protein estrogen receptor GPER/GPR30 mediates estrogen action as the cardioprotection under certain stressful conditions. For instance, GPER activation by the selective agonist G-1 reduced myocardial inflammation, improved immunosuppression, triggered pro-survival signaling cascades, improved myocardial mechanical performance, and reduced infarct size after ischemia/reperfusion (I/R) injury. Hence, we evaluated whether ligand-activated GPER may exert cardioprotection in male rats chronically treated with Dox. 1 week of G-1 (50 µg/kg/day) intraperitoneal administration mitigated Dox (3 mg/kg/day) adverse effects, as revealed by reduced TNF-α, IL-1ß, LDH, and ROS levels. Western blotting analysis of cardiac homogenates indicated that G-1 prevents the increase in p-c-jun, BAX, CTGF, iNOS, and COX2 expression induced by Dox. Moreover, the activation of GPER rescued the inhibitory action elicited by Dox on the expression of BCL2, pERK, and pAKT. TUNEL assay indicated that GPER activation may also attenuate the cardiomyocyte apoptosis upon Dox exposure. Using ex vivo Langendorff perfused heart technique, we also found an increased systolic recovery and a reduction of both infarct size and LDH levels in rats treated with G-1 in combination with Dox respect to animals treated with Dox alone. Accordingly, the beneficial effects induced by G-1 were abrogated in the presence of the GPER selective antagonist G15. These data suggest that GPER activation mitigates Dox-induced cardiotoxicity, thus proposing GPER as a novel pharmacological target to limit the detrimental cardiac effects of Dox treatment. J. Cell. Physiol. 232: 1640-1649, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Doxorubicin/adverse effects , Quinolines/therapeutic use , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Biomarkers/metabolism , Blood Pressure/drug effects , Cardiotonic Agents/pharmacology , Cardiotoxicity/blood , Cardiotoxicity/pathology , Cardiotoxicity/physiopathology , Diastole/drug effects , Heart Function Tests/drug effects , Humans , Inflammation/pathology , Interleukin-1beta/blood , L-Lactate Dehydrogenase/blood , Ligands , Male , Myocardial Ischemia/blood , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Quinolines/pharmacology , Rats, Wistar , Reactive Oxygen Species/blood , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Tumor Necrosis Factor-alpha/blood , Ventricular Function/drug effects
6.
Front Physiol ; 7: 610, 2016.
Article in English | MEDLINE | ID: mdl-28082904

ABSTRACT

Magnesium chloride (MgCl2) is commonly used as a general anesthetic in cephalopods, but its physiological effects including those at cardiac level are not well-characterized. We used an in vitro isolated perfused systemic heart preparation from the common octopus, Octopus vulgaris, to investigate: (a) if in vivo exposure to MgCl2 formulations had an effect on cardiac function in vitro and, if so, could this impact recovery and (b) direct effects of MgCl2 formulations on cardiac function. In vitro hearts removed from animals exposed in vivo to 3.5% MgCl2 in sea water (20 min) or to a mixture of MgCl2+ ethanol (1.12/1%; 20 min) showed cardiac function (heart rate, stroke volume, cardiac output) comparable to hearts removed from animals killed under hypothermia. However, 3.5% MgCl2 (1:1, sea water: distilled water, 20 min) produced a significant impairment of the Frank-Starling response as did 45 min exposure to the MgCl2+ ethanol mixture. Perfusion of the isolated heart with MgCl2± ethanol formulations produced a concentration-related bradycardia (and arrest), a decreased stroke volume and cardiac output indicating a direct effect on the heart. The cardiac effects of MgCl2 are discussed in relation to the involvement of magnesium, sodium, chloride, and calcium ions, exposure time and osmolality of the formulations and the implications for the use of various formulations of MgCl2 as anesthetics in octopus. Overall, provided that the in vivo exposure to 3.5% MgCl2 in sea water or to a mixture of MgCl2+ ethanol is limited to ~20 min, residual effects on cardiac function are unlikely to impact post-anesthetic recovery.

7.
Eur J Pharm Sci ; 82: 161-70, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26631584

ABSTRACT

The antihypertensive flavonol quercetin (Q1) is endowed with a cardioprotective effect against myocardial ischemic damage. Q1 inhibits angiotensin converting enzyme activity, improves vascular relaxation, and decreases oxidative stress and gene expression. However, the clinical application of this flavonol is limited by its poor bioavailability and low stability in aqueous medium. In the aim to overcome these drawbacks and preserve the cardioprotective effects of quercetin, the present study reports on the preparation of five different Q1 analogs, in which all OH groups were replaced by hydrophobic functional moieties. Q1 derivatives have been synthesized by optimizing previously reported procedures and analyzed by spectroscopic analysis. The cardiovascular properties of the obtained compounds were also investigated in order to evaluate whether chemical modification affects their biological efficacy. The interaction with ß-adrenergic receptors was evaluated by molecular docking and the cardiovascular efficacy was investigated on the ex vivo Langendorff perfused rat heart. Furthermore, the bioavailability and the antihypertensive properties of the most active derivative were evaluated by in vitro studies and in vivo administration (1month) on spontaneously hypertensive rats (SHRs), respectively. Among all studied Q1 derivatives, only the ethyl derivative reduced left ventricular pressure (at 10(-8)M÷10(-6)M doses) and improved relaxation and coronary dilation. NOSs inhibition by L-NAME abolished inotropism, lusitropism and coronary effects. Chronic administration of high doses of this compound on SHR reduced systolic and diastolic pressure. Differently, the acetyl derivative induced negative inotropism and lusitropism (at 10(-10)M and 10(-8)÷10(-6)M doses), without affecting coronary pressure. Accordingly, docking studies suggested that these compounds bind both ß1/ß2-adrenergic receptors. Taking into consideration all the obtained results, the replacement of OH with ethyl groups seems to improve Q1 bioavailability and stability; therefore, the ethyl derivative could represent a good candidate for clinical use in hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Quercetin/pharmacology , Animals , Antihypertensive Agents/chemistry , Biological Availability , Dose-Response Relationship, Drug , Gastric Juice/metabolism , Heart/drug effects , Heart/physiology , Intestinal Secretions/metabolism , Models, Molecular , Pancreatin/metabolism , Pepsin A/metabolism , Quercetin/analogs & derivatives , Quercetin/chemistry , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism
8.
Curr Drug Targets ; 16(8): 877-83, 2015.
Article in English | MEDLINE | ID: mdl-25850621

ABSTRACT

Recently, the hypothalamic 82-aa peptide Nesfatin-1 received notable attention for its anorexigenic and anti-hyperglycemic properties. In mammalian hypothalamus, Nesfatin-1 is expressed, together with the precursor Nucleobindin 2 (NUCB2), in regions controlling water-food intake, body weight, and glucose homeostasis. The peptide is also peripherally expressed, as shown in the rat heart, in which it is present together with NUCB2. In addition to a central modulation of nutrition and energy balance, and of the nervous circuits responsible for blood pressure and heart rate control, Nesfatin-1 also acts peripherally on several districts, including the cardiovascular (CV) system. Accordingly, the peptide is regarded with interest as a multifunctional hormone not only linked to alimentary homeostasis. This review aims to analyze the literature on Nesfatin-1, with focus on its emerging CV activity. Few available studies show that the peptide affects energy metabolism of murine and human cardiomyocytes, by eliciting insulin-like effects. On the ex vivo rat heart, it directly depresses contractility and relaxation via cGMP, PKG and ERK1/2, and limits ischemia/reperfusion (I/R) damage, acting in post-conditioning protection. Nesfatin-1 actions are proposed to involve an unknown G-protein coupled receptor. However, in the rat heart, functional studies, co-immunoprecipitation and local sequence alignment analyses suggest an interaction with the Natriuretic Peptide Receptor-type A (NPR-A). These data open up novel perspectives to clarify not only the biological significance of the peptide, but also its putative biomedical potential in the presence of nutrition-dependent cardiovascular diseases.


Subject(s)
Calcium-Binding Proteins/metabolism , Cardiotonic Agents/metabolism , DNA-Binding Proteins/metabolism , Myocardium/metabolism , Nerve Tissue Proteins/metabolism , Animals , Calcium-Binding Proteins/genetics , DNA-Binding Proteins/genetics , Energy Metabolism , Humans , Ischemic Postconditioning/methods , Nerve Tissue Proteins/genetics , Nucleobindins , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction
9.
Front Chem ; 2: 64, 2014.
Article in English | MEDLINE | ID: mdl-25177680

ABSTRACT

Together with Chromogranin B and Secretogranins, Chromogranin A (CGA) is stored in secretory (chromaffin) granules of the diffuse neuroendocrine system and released with noradrenalin and adrenalin. Co-stored within the granule together with neuropeptideY, cardiac natriuretic peptide hormones, several prohormones and their proteolytic enzymes, CGA is a multifunctional protein and a major marker of the sympatho-adrenal neuroendocrine activity. Due to its partial processing to several biologically active peptides, CGA appears an important pro-hormone implicated in relevant modulatory actions on endocrine, cardiovascular, metabolic, and immune systems through both direct and indirect sympatho-adrenergic interactions. As a part of this scenario, we here illustrate the emerging role exerted by the full-length CGA and its three derived fragments, i.e., Vasostatin 1, catestatin and serpinin, in the control of circulatory homeostasis with particular emphasis on their cardio-vascular actions under both physiological and physio-pathological conditions. The Vasostatin 1- and catestatin-induced cardiodepressive influences are achieved through anti-beta-adrenergic-NO-cGMP signaling, while serpinin acts like beta1-adrenergic agonist through AD-cAMP-independent NO signaling. On the whole, these actions contribute to widen our knowledge regarding the sympatho-chromaffin control of the cardiovascular system and its highly integrated "whip-brake" networks.

10.
PLoS One ; 9(8): e102536, 2014.
Article in English | MEDLINE | ID: mdl-25099124

ABSTRACT

BACKGROUND: In the presence of comorbidities the effectiveness of many cardioprotective strategies is blunted. The goal of this study was to assess in a hypertensive rat model if the early reperfusion with anti-hypertensive and pro-angiogenic Chromogranin A-derived peptide, Catestatin (CST:hCgA352-372; CST-Post), protects the heart via Reperfusion-Injury-Salvage-Kinases (RISK)-pathway activation, limiting infarct-size and apoptosis, and promoting angiogenetic factors (e.g., hypoxia inducible factor, HIF-1α, and endothelial nitric oxide synthase, eNOS, expression). METHODS AND RESULTS: The effects of CST-Post on infarct-size, apoptosis and pro-angiogenetic factors were studied in isolated hearts of spontaneously hypertensive rats (SHR), which underwent the following protocols: (a) 30-min ischemia and 120-min reperfusion (I/R); (b) 30-min ischemia and 20-min reperfusion (I/R-short), both with and without CST-Post (75 nM for 20-min at the beginning of reperfusion). In unprotected Wistar-Kyoto hearts, used as normal counterpart, infarct-size resulted smaller than in SHR. CST-Post reduced significantly infarct-size and improved post-ischemic cardiac function in both strains. After 20-min reperfusion, CST-Post induced S-nitrosylation of calcium channels and phosphorylation of RISK-pathway in WKY and SHR hearts. Yet specific inhibitors of the RISK pathway blocked the CST-Post protective effects against infarct in the 120-min reperfusion groups. Moreover, apoptosis (evaluated by TUNEL, ARC and cleaved caspase) was reduced by CST-Post. Importantly, CST-Post increased expression of pro-angiogenetic factors (i.e., HIF-1α and eNOS expression) after two-hour reperfusion. CONCLUSIONS: CST-Post limits reperfusion damages and reverses the hypertension-induced increase of I/R susceptibility. Moreover, CST-Post triggers antiapoptotic and pro-angiogenetic factors suggesting that CST-Post can be used as an anti-maladaptive remodeling treatment.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Cardiomegaly/metabolism , Chromogranin A/pharmacology , Gene Expression Regulation/drug effects , Myocardial Ischemia/metabolism , Myocardium/metabolism , Peptide Fragments/pharmacology , Animals , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Disease Models, Animal , Female , Male , Myocardial Ischemia/drug therapy , Myocardial Ischemia/pathology , Myocardium/pathology , Rats , Rats, Inbred SHR
11.
FASEB J ; 26(7): 2888-98, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22459152

ABSTRACT

Three forms of serpinin peptides, serpinin (Ala26Leu), pyroglutaminated (pGlu)-serpinin (pGlu23Leu), and serpinin-Arg-Arg-Gly (Ala29Gly), are derived from cleavage at pairs of basic residues in the highly conserved C terminus of chromogranin A (CgA). Serpinin induces PN-1 expression in neuroendocrine cells to up-regulate granule biogenesis via a cAMP-protein kinase A-Sp1 pathway, while pGlu-serpinin inhibits cell death. The aim of this study was to test the hypothesis that serpinin peptides are produced in the heart and act as novel ß-adrenergic-like cardiac modulators. We detected serpinin peptides in the rat heart by HPLC and ELISA methods. The peptides included predominantly Ala29Gly and pGlu-serpinin and a small amount of serpinin. Using the Langendorff perfused rat heart to evaluate the hemodynamic changes, we found that serpinin and pGlu-serpinin exert dose-dependent positive inotropic and lusitropic effects at 11-165 nM, within the first 5 min after administration. The pGlu-serpinin-induced contractility is more potent than that of serpinin, starting from 1 nM. Using the isolated rat papillary muscle preparation to measure contractility in terms of tension development and muscle length, we further corroborated the pGlu-serpinin-induced positive inotropism. Ala29Gly was unable to affect myocardial performance. Both pGlu-serpinin and serpinin act through a ß1-adrenergic receptor/adenylate cyclase/cAMP/PKA pathway, indicating that, contrary to the ß-blocking profile of the other CgA-derived cardiosuppressive peptides, vasostatin-1 and catestatin, these two C-terminal peptides act as ß-adrenergic-like agonists. In cardiac tissue extracts, pGlu-serpinin increased intracellular cAMP levels and phosphorylation of phospholamban (PLN)Ser16, ERK1/2, and GSK-3ß. Serpinin and pGlu-serpinin peptides emerge as novel ß-adrenergic inotropic and lusitropic modulators, suggesting that CgA and the other derived cardioactive peptides can play a key role in how the myocardium orchestrates its complex response to sympathochromaffin stimulation.


Subject(s)
Adrenergic beta-1 Receptor Agonists/chemistry , Adrenergic beta-1 Receptor Agonists/pharmacology , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Chromogranin A/chemistry , Chromogranin A/physiology , Heart/drug effects , Heart/physiology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Chromogranin A/genetics , Chromogranin A/pharmacology , In Vitro Techniques , Male , Molecular Sequence Data , Myocardial Contraction/drug effects , Myocardium/chemistry , Papillary Muscles/drug effects , Papillary Muscles/physiology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Rats , Rats, Wistar
12.
J Nutr Biochem ; 23(1): 69-75, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21414764

ABSTRACT

Root extract of liquorice is traditionally used to treat several diseases. Liquorice-derived constituents present several biological actions. In particular, glycyrrhizin and its aglycone, glycyrrhetinic acid, exhibit well-known cardiovascular properties. The aim of this research was to explore the direct cardiac activity of glycyrrhizin and glycyrrhetinic acid. The effects of synthetic glycyrrhizin and glycyrrhetinic acid were evaluated on the isolated and Langendorff perfused rat heart. The intracellular signaling involved in the effects of the two substances was analyzed on isolated and perfused heart and by Western blotting on cardiac extracts. Under basal conditions, both glycyrrhizin and glycyrrhetinic acid influenced cardiac contractility and relaxation. Glycyrrhizin induced significant positive inotropic and lusitropic effects starting from very low concentrations, while both inotropism and lusitropism were negatively affected by glycyrrhetinic acid. Both substances significantly increased heart rate. Analysis of the signal transduction mechanisms suggested that glycyrrhizin acts through the endothelin receptor type A/phospholipase C axis while glycyrrhetinic acid acts through endothelin receptor type B/Akt/nitric oxide synthase/nitric oxide axis. To our knowledge, these data reveal, for the first time, that both glycyrrhizin and glycyrrhetinic acid directly affect cardiac performance. Additional information on the physiological significance of these substances and their cardiac molecular targets may provide indication on their biomedical application.


Subject(s)
Glycyrrhetinic Acid/pharmacology , Glycyrrhizic Acid/pharmacology , Heart/drug effects , Animals , Dose-Response Relationship, Drug , Heart Rate/drug effects , In Vitro Techniques , Myocardial Contraction/drug effects , Myocardium/metabolism , Perfusion , Rats , Receptor, Endothelin A/metabolism , Signal Transduction , Type C Phospholipases/metabolism
13.
Cell Physiol Biochem ; 28(1): 41-52, 2011.
Article in English | MEDLINE | ID: mdl-21865847

ABSTRACT

BACKGROUND/AIMS: 17ß-estradiol (17ßE2) plays an important cardiovascular role by activating estrogen receptors (ER) α and ERß. Previous studies demonstrated that the novel estrogen G protein-coupled receptor (GPR30/GPER) mediates estrogen action in different tissues. We have recently shown in the rat heart that 17ßE2 elicits negative inotropism through ERα, ERß and GPR30, by triggering activation of ERK1/2, phosphatidylinositol 3-kinase (PI3K), protein kinase A (PKA) and endothelial Nitric Oxide synthase (eNOS) signaling. METHODS: In the present study, using the isolated and Langendorff-perfused rat heart as a model system we analyzed: i) whether and to which extent 17ßE2 modifies mammalian ventricular myocardial relaxation (lusitropism); ii) the type of ERs and the signaling pathways involved in this effect. RESULTS: We found that 17ßE2 negatively modulated the ventricular lusitropic performance. This effect, which partially involved the vascular endothelium, recruited ERß and occurred via PI3K, eNOS-NO-cGMP-protein kinase G (PKG) transduction cascade. Of note, 17ßE2-mediated negative lusitropism associated with a modification of phospholamban (PLN) phosphorylation and S-nitrosylation (SNO) both in isolated Langendorff rat heart and in isolated cardiomyocytes. CONCLUSION: Taken together, our results allow including 17ßE2 to the family of substances that control ventricular relaxation. This is of relevance in relation not only to the normal endocrine control of cardiac function, but also to physio-pathologic conditions characterized by an altered ventricular diastolic performance.


Subject(s)
Calcium-Binding Proteins/metabolism , Estradiol/pharmacology , Heart Ventricles/drug effects , Animals , Cyclic GMP-Dependent Protein Kinases/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Heart Ventricles/metabolism , In Vitro Techniques , Male , Muscle Contraction , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Signal Transduction/drug effects
14.
Nitric Oxide ; 20(2): 69-78, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19027084

ABSTRACT

The nitric oxide synthase (NOS)/nitric oxide (NO) system integrates cellular biochemical machinery and energetics. In heart microenvironment, dynamic NO behaviour depends upon the presence of superoxide anions, haemoglobin (Hb), and myoglobin (Mb), being hemoproteins are major players disarming NO bioactivity. The Antarctic icefish, which lack Hb and, in some species, also cardiac Mb, represent a unique model for exploring Hb and Mb impact on NOS/NO function. We report in the (Hb(-)/Mb(-)) icefish Chaenocephalus aceratus the presence of cardiac NOSs activity (NADPH-diaphorase) and endothelial NOS (eNOS)/inducible NOS (iNOS) zonal immuno-localization in the myocardium. eNOS is localized on endocardium and, to a lesser extent, in myocardiocytes, while iNOS is localized exclusively in myocardiocytes. Confronting eNOS and iNOS expression in Trematomus bernacchii (Hb(+)/Mb(+)), C. hamatus (Hb(-)/Mb(+)) and C. aceratus (Hb(-)/Mb(-)) is evident a lower expression in the Mb-less icefish. NO signaling was analyzed using isolated working heart preparations. In T. bernacchii, L-arginine and exogenous (SIN-1) NO donor dose-dependently decreased stroke volume, indicating decreased inotropism. L-arginine-induced inotropism was NOSs-dependent, being abolished by NOSs-inhibitor NG-monomethyl-L-arginine (L-NMMA). A SIN-1-induced negative inotropism was found in presence of SOD. NOS inhibition by L-N5-N-iminoethyl-L-ornithine (L-NIO) and L-NMMA confirmed the NO-mediated negative inotropic influence on cardiac performance. In contrast, in C. aceratus, L-arginine elicited a positive inotropism. SIN-1 induced a negative inotropism, which disappeared in presence of SOD, indicating peroxynitrite involvement. Cardiac performance was unaffected by L-NIO and L-NIL. NO signaling acted via a cGMP-independent mechanism. This high conservation degree of NOS localization pattern and signaling highlights its importance for cardiac biology.


Subject(s)
Myocardium/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Perciformes/metabolism , Animals , Antarctic Regions , Arginine/metabolism , Blotting, Western , Cyclic GMP/metabolism , Endocardium/metabolism , Female , Fluorescent Antibody Technique , Heart Ventricles/metabolism , Hemodynamics , Male , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , NADPH Dehydrogenase/metabolism , Signal Transduction , Stroke Volume , omega-N-Methylarginine/pharmacology
15.
J Anat ; 213(2): 106-19, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18482286

ABSTRACT

This paper reports on the structure and ultrastructure of the ventricular myocardium of the African lungfish Protopterus dolloi in freshwater (FW), in aestivation (AE), and after the AE period. The myocardium shows a conventional myofibrillar structure. All the myocytes contain large intracytoplasmic spaces occupied by a pale material that could contain glycosaminoglycans and/or glycogen, which may be used as food and water reservoirs. In FW, the myocytes in the trabeculae associated with the free ventricular wall show structural signs of low transcriptional and metabolic activity (heterochromatin, mitochondria of the dense type). These signs are partially reversed during the AE period (euchromatin, mitochondria with a light matrix), with a return to the FW appearance after arousal. The myocytes in the septum show, in FW conditions, nuclear polymorphism (heterochromatin, euchromatin), and two types (colliquative and coagulative) of necrosis. In AE, all the septal myocytes show euchromatin, and the number of necrotic cells increases greatly. Cell necrosis appears to be related to the septal architecture. After arousal, the septal myocytes exhibit a heterochromatin pattern, the number of necrotic cells decreases, cell debris accumulates under the endocardium, and phagocytosis takes place. Despite being a morphologic continuum, the trabeculae associated with the free ventricular wall appear to constitute a different compartment from that formed by the trabeculae in the ventricular septum. Paradoxically, AE appears to trigger an increase in transcriptional and synthetic myocardial activities, especially at the level of the ventricular septum. This activity may be involved in mechanisms of autocrine/paracrine regulation. Aestivation cannot be regarded as the result of a general depression of all cellular and organic activities. Rather, it is a much more complex state in which the interplay between upregulation and downregulation of diverse cell activities appears to play a fundamental role.


Subject(s)
Estivation/physiology , Fishes/anatomy & histology , Myocardium/ultrastructure , Animals , Fishes/metabolism , Fishes/physiology , Fresh Water , Heart Ventricles/metabolism , Heart Ventricles/ultrastructure , Microscopy, Electron , Microscopy, Electron, Scanning , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Ventricular Septum/metabolism , Ventricular Septum/ultrastructure
16.
Anat Rec ; 268(4): 388-98, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12420287

ABSTRACT

Sturgeons constitute a family of living "fossil" fish whose heart is related to that of other ancient fish and the elasmobranches. We have undertaken a systematic study of the structure of the sturgeon heart aimed at unraveling the relationship between the heart structure and the adaptive evolutionary changes. In a related paper, data were presented on the conus valves and the subendocardium. Here, the structure of the conus myocardium, the subepicardial tissue, and the conus-aorta transition were studied by conventional light, transmission, and scanning electron microscopy. In addition, actin localization by fluorescent phalloidin was used. The conus myocardium is organized into bundles whose spatial organization changes along the conus length. The variable orientation of the myocardial cell bundles may be effective in emptying the conus lumen during contraction and in preventing reflux of blood. Myocardial cell bundles are separated by loose connective tissue that contains collagen and elastin fibers, vessels, and extremely flat cells separating the cell bundles and enclosing blood vessels and collagen fibers. The ultrastructure of the myocardial cells was found to be very similar to that of other fish groups, suggesting that it is largely conservative. The subepicardium is characterized by the presence of nodular structures that contain lympho-hemopoietic (thymus-like) tissue in the young sturgeons and a large number of lymphocytes after the sturgeons reach sexual maturity. This tissue is likely implicated in the establishment and maintenance of the immune responses. The intrapericardial ventral aorta shows a middle layer of circumferentially oriented cells and internal and external layers with cells oriented longitudinally. Elastin fibers completely surround each smooth muscle cell, and the spaces between the different layers are occupied by randomly arranged collagen bundles. The intrapericardial segment of the ventral aorta is a true transitional segment whose structural characteristics are different from those of both the conus subendocardium and the rest of the ventral aorta.


Subject(s)
Aorta/ultrastructure , Fishes/anatomy & histology , Myocardium/ultrastructure , Pericardium/ultrastructure , Truncus Arteriosus/ultrastructure , Animals , Lymphoid Tissue/ultrastructure , Microscopy, Electron
17.
Anat Rec ; 267(1): 17-27, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11984788

ABSTRACT

Sturgeons are bony fish that retain structural traits typical of the more primitive Chondrostei. From an evolutionary viewpoint, sturgeons are considered relic fish. However, they show remarkable ecological plasticity and are well adapted to contemporary environmental conditions. Although development of the cardiovascular system is critical for all organs and systems, and is affected by evolutionary changes, the structure of the sturgeon heart has been mostly overlooked. This is also true for the conus arteriosus, which, as in Chondrostei, is endowed with several rows of valves and a layer of contractile myocardium. This work reports on the structure of the valves, the endocardium, and the subendocardium of the conus arteriosus of the sturgeon (Acipenser naccarii) heart. It is part of a broader study that aims to cover the entire structure of the sturgeon heart. The conus arteriosus of 15 A. naccarii hearts, ranging in age from juveniles to sexually-differentiated adults, has been studied by conventional light, transmission (TEM), and scanning electron microscopy (SEM). In addition, maceration of the soft tissues with NaOH, and actin localization by fluorescent phalloidin has been used. The conus is a tubular chamber that arises from the right ventricular side and presents two constrictions at the conus-ventricle and conus-aorta junctions. The conus is endowed with three rows of valves: one distal and two proximal. The segment of the conus located between the distal and the two proximal rows is devoid of valvular structures. The distal row has four leaflets, while the two proximal rows show the greatest variation in leaflet number, size, and shape. All leaflets have collagenous chordae tendineae arising from the free border and from the parietal side of the leaflets. The endocardium is a flat endothelium which shows a thick, irregular basement membrane. The leaflet body is formed by a loose connective tissue which blends with the subendocardium. The subendocardium is a connective tissue consisting of myofibroblasts, collagen, and elastin. It is divided into two distinct areas: one proximal, which shows little elastin and poorly organized collagen; and one distal, which is rich in elastin, with cells and extracellular fibers organized into layers that are oriented in alternative circumferential and longitudinal directions. The present report is the first systematic analysis of the structure of the sturgeon conus. Descriptions of the conus valves should recognize the existence of three valve rows only. The variability in valve morphology, and the loose structure of the leaflet tissue make it unlikely that the valves play an effective role in preventing blood backflow. In this regard, the ventricle-conus constriction may act as a sphincter. The subendocardium is an elastic coat capable of actively sustaining the tissue deformation that accompanies the heart contractile cycle. Further comparative studies are needed to provide deeper insight into the structural changes that accompany phyletic diversification.


Subject(s)
Endocardium/anatomy & histology , Fishes/anatomy & histology , Heart Valves/anatomy & histology , Truncus Arteriosus/anatomy & histology , Animals , Endocardium/growth & development , Fishes/growth & development , Heart Valves/growth & development , Microscopy, Electron, Scanning , Truncus Arteriosus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...