Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Acta Physiol (Oxf) ; 223(4): e13067, 2018 08.
Article in English | MEDLINE | ID: mdl-29575758

ABSTRACT

AIM: Selenoprotein T (SelT or SELENOT) is a novel thioredoxin-like enzyme whose genetic ablation in mice results in early embryonic lethality. SelT exerts an essential cytoprotective action during development and after injury through its redox-active catalytic site. This study aimed to determine the expression and regulation of SelT in the mammalian heart in normal and pathological conditions and to evaluate the cardioprotective effect of a SelT-derived peptide, SelT43-52(PSELT) encompassing the redox motif which is key to its function, against ischaemia/reperfusion(I/R) injury. METHODS: We used the isolated Langendorff rat heart model and different analyses by immunohistochemistry, Western blot and ELISA. RESULTS: We found that SelT expression is very abundant in embryo but is undetectable in adult heart. However, SelT expression was tremendously increased after I/R. PSELT (5 nmol/L) was able to induce pharmacological post-conditioning cardioprotection as evidenced by a significant recovery of contractility (dLVP) and reduction of infarct size (IS), without changes in cardiac contracture (LVEDP). In contrast, a control peptide lacking the redox site did not confer cardioprotection. Immunoblot analysis showed that PSELT-dependent cardioprotection is accompanied by a significant increase in phosphorylated Akt, Erk-1/2 and Gsk3α-ß, and a decrement of p38MAPK. PSELT inhibited the pro-apoptotic factors Bax, caspase 3 and cytochrome c and stimulated the anti-apoptotic factor Bcl-2. Furthermore, PSELT significantly reduced several markers of I/R-induced oxidative and nitrosative stress. CONCLUSION: These results unravel the role of SelT as a cardiac modulator and identify PSELT as an effective pharmacological post-conditioning agent able to protect the heart after ischaemic injury.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Selenoproteins/pharmacology , Thioredoxin-Disulfide Reductase/pharmacology , Animals , Antioxidants/metabolism , Apoptosis Regulatory Proteins/metabolism , Disease Models, Animal , Isolated Heart Preparation , Male , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nitrosative Stress/drug effects , Peptide Fragments/metabolism , Rats, Wistar , Selenoproteins/metabolism , Signal Transduction/drug effects , Thioredoxin-Disulfide Reductase/metabolism , Ventricular Function, Left/drug effects
2.
Gen Comp Endocrinol ; 240: 1-9, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27633326

ABSTRACT

Chromogranin A (CgA) is an acidic protein co-stored with catecholamines, hormones and neuropeptides in the secretory granules of endocrine, neuronal and other cell types (including cardiomyocytes). Proteolytic cleavage in the C terminus of CgA generates a 2.9kDa peptide named serpinin (Serp; Ala26Leu) that can be modified at its N terminus to form a pyroglutamate residue (pGlu-Serp). In the rat heart, both peptides increase contractility and relaxation through a ß-adrenergic-like action mechanism. Accordingly, Serp and pGlu-Serp were proposed as novel myocardial sympatho-adrenergic modulators in mammals. On a comparative basis, here we report the actions of Serp and pGlu-Serp on myocardial contractility in three poikilotherm vertebrate species: the eel (Anguilla anguilla), the goldfish (Carassius auratus) and the frog (Rana esculenta). Using isolated working heart preparations, we show that pGlu-Serp reduces stroke volume in all species tested, while Serp reduces contractility in the frog heart, but is uneffective in eel and goldfish hearts. In the goldfish and frog hearts, pGlu-Serp activates the Nitric Oxide/cGMP pathway involving Endothelin-1 B receptors (frog) and ß3 adrenergic receptors (goldfish). pGlu-Serp-treated hearts from goldfish and frog show increased cGMP content. Moreover, the exposure of the frog heart to pGlu-Serp is accompanied by an increased expression of activated eNOS and Akt. In conclusion, this first report showing that pGlu-Serp inhibits mechanical cardiac performance in teleost and amphibians supports an evolutionary role of the CgA system, and particularly its serpinin component, in the sympatho-adrenergic control of the vertebrate heart.


Subject(s)
Amphibians/metabolism , Chromogranin A/genetics , Heart/drug effects , Myocardium/metabolism , Sympathomimetics/metabolism , Animals , Peptide Fragments/metabolism , Signal Transduction
3.
J Endocrinol ; 227(3): 167-178, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26400960

ABSTRACT

Serpinin peptides derive from proteolytic cleavage of Chromogranin-A at C-terminus. Serpinin and the more potent pyroglutaminated-serpinin (pGlu-Serp) are positive cardiac ß-adrenergic-like modulators, acting through ß1-AR/AC/cAMP/PKA pathway. Because in some conditions this pathway and/or other pro-survival pathways, activated by other Chromogranin-A fragments, may cross-talk and may be protective, here we explored whether pGlu-Serp cardioprotects against ischemia/reperfusion injury under normotensive and hypertensive conditions. In the latter condition, cardioprotection is often blunted because of the limitations on pro-survival Reperfusion Injury Salvage Kinases (RISK) pathway activation. The effects of pGlu-Serp were evaluated on infarct size (IS) and cardiac function by using the isolated and Langendorff perfused heart of normotensive (Wistar Kyoto, WKY) and spontaneously hypertensive (SHR) rats exposed to ischemic pre-conditioning (PreC) and post-conditioning (PostC). In both WKY and SHR rat, pGlu-Serp induced mild cardioprotection in both PreC and PostC. pGlu-Serp administered at the reperfusion (Serp-PostC) significantly reduced IS, being more protective in SHR than in WKY. Conversely, left ventricular developed pressure (LVDevP) post-ischemic recovery was greater in WKY than in SHR. pGlu-Serp-PostC reduced contracture in both strains. Co-infusion with specific RISK inhibitors (PI3K/Akt, MitoKATP channels and PKC) blocked the pGlu-Serp-PostC protective effects. To show direct effect on cardiomyocytes, we pre-treated H9c2 cells with pGlu-Serp, which were thus protected against hypoxia/reoxygenation. These results suggest pGlu-Serp as a potential modulatory agent implicated in the protective processes that can limit infarct size and overcome the hypertension-induced failure of PostC.


Subject(s)
Chromogranin A/therapeutic use , Hypertension/complications , Ischemic Preconditioning, Myocardial/methods , Myocardial Ischemia/prevention & control , Peptide Fragments/therapeutic use , Animals , Chromogranin A/pharmacology , Enzyme Inhibitors/pharmacology , Hypertension/metabolism , Male , Myocardial Ischemia/complications , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Myocardium/metabolism , Peptide Fragments/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects
4.
Nitric Oxide ; 44: 71-80, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25499100

ABSTRACT

African lungfishes are obligate air breathers, with reduced gills and pulmonary breathing throughout their life. During the dry season they aestivate on land, with the collapse of secondary lamellae of their gills and the establishment of an exclusive aerial ventilation through the vascularization and expansion of their lungs. To date, the mechanisms underlining the respiratory organ remodeling in aestivating lungfishes are unknown. This study aimed to identify key switch components of the stress-induced signal transduction networks implicated in both rapid and medium-long term remodeling of the gills and lungs of the African lungfish Protopterus annectens during aestivation. Through immunofluorescence microscopy and Western blotting, the localization and the expression of nitric oxide synthase (NOS), Akt, Hsp-90 and HIF-1α were evaluated in both gills and lungs exposed to three experimental conditions: freshwater (FW), 6 months of experimentally induced aestivation (6mAe), and 6 days after arousal from 6 months of aestivation (6mAe6d). After 6mAe, the expression of NOS (p-eNOS antibody), Akt (p-Akt antibody), and Hsp-90 decreased in the gills, while NOS and Hsp-90 expression increased with Akt remained unchanged in the lungs. Upon 6mAe6d, NOS, Akt and Hsp-90 expression in the gills returned to the respective FW values. In the lungs of the aroused fish, NOS and Akt decreased to their respective FW levels, while Hsp-90 expression was enhanced with respect to aestivation. In both respiratory organs, the qualitative and quantitative patterns of HIF-1α expression correlated inversely to those of NOS. Overall, our findings suggest that the molecular components of the NOS/NO system changed in a tissue-specific manner in parallel with organ readjustment in the gills and lungs of P. annectens during aestivation and arousal.


Subject(s)
Estivation/physiology , Gills/chemistry , Lung/chemistry , Nitric Oxide Synthase/analysis , Signal Transduction/physiology , Animals , Blotting, Western , Fishes , Gills/metabolism , Lung/metabolism , Nitric Oxide Synthase/metabolism
5.
Curr Med Chem ; 22(3): 292-304, 2015.
Article in English | MEDLINE | ID: mdl-25386823

ABSTRACT

Catestatin (CST), the Chromogranin A (CgA)-derived cationic and hydrophobic peptide, firstly recognized as an endogenous inhibitor of catecholamine secretion, functions as a physiological brake of the adreno-sympathetic-chromaffin system. Its wide spectrum of activities includes relevant multilevel cardiovascular and antihypertensive influences. At central systemic level, CST seems to modulate the autonomic cardiovascular control possibly acting on baroreceptor afferent fibers of the nucleus tractus solitarius. This, as well as clinical and experimental (CgA-KO mice) evidences point to an important role of CST in the determinism and prevention of essential hypertension. At organ level, CST exerts myocardial (negative inotropy and lusitropy) effects and potently vasodilates endothelin-1 (ET-1)-preconstricted coronaries through ß2-adrenergic receptor (AR)-Gi/o protein-nitric oxide (NO)-cGMP signalling, while counterbalancing ß adrenergic (ISO) stimulation. The contractile myocardial effects have been deeply analysed in fish and amphibian hearts, highlighting finely diversified mechanisms of action. CST also acts as cardioprotective agent in both pre- and post-conditioning through NO-dependent mechanisms implicating the Reperfusion Injury Salvage Kinase (RISK) signalling and the activation of mitoKATP channels. The CST-elicited cardiotropic and coronarotropic influences, along with the recently discovered proangiogenic and regulatory effects in glucose and lipid metabolism, contribute to delineate an integrated and updated picture of the peptide which emerges as a pleiotropic hormone with a wide range of cytokine-like characteristics. The aim of this review is to interlock some older and more recent evidences which may help to better perceive the subtle links and differences among the puzzle pieces that still need to be deciphered.


Subject(s)
Cardiotonic Agents/pharmacology , Cardiovascular System/drug effects , Chromogranin A/pharmacology , Peptide Fragments/pharmacology , Animals , Cardiovascular System/metabolism , Coronary Vessels/drug effects , Coronary Vessels/physiology , Heart/drug effects , Heart/physiology , Humans , Neovascularization, Physiologic/drug effects
6.
Nitric Oxide ; 42: 24-31, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25178168

ABSTRACT

Goldfish tolerate prolonged and severe hypoxia, thus representing a well-suited model to study the maintenance of cardiac function when O(2) availability represents a limiting factor. Using a working heart preparation, we explored the role of the intracardiac nitric oxide synthase (NOS)-derived nitric oxide (NO) under normoxic and hypoxic conditions. Cardiac performance was examined both under basal (constant preload and afterload) and loading conditions, i.e. preload-induced increases in stroke volume (SV) and hence cardiac output at constant afterload (the Frank-Starling response). Hypoxic hearts showed an increased basal mechanical performance compared to the normoxic counterpart. Under basal conditions, in both normoxic and hypoxic hearts, NOS and soluble guanylyl cyclase (sGC) inhibition increased SV, while exogenous NO supply decreased it. The normoxic heart was very sensitive to filling pressure increases; the maximum SV = 1.08 ± 0.09 mL/kg body mass was obtained at 0.4 kPa. Acute hypoxia increased this sensitivity, SV reaching the maximum value (1.45 ± 0.12 mL/kg body mass) at 0.25 kPa. NOS inhibition by L-NMMA reduced the Frank-Starling response under normoxia, but was ineffective under acute hypoxia, where NO may come from nitrite reduction. In both conditions, sGC inhibition induced a reduction of the cardiac response to preload. Moreover, under acute hypoxia, NO scavenging significantly reduced the Frank-Starling response. The hypoxia-induced hemodynamic patterns were complemented by Western blotting analysis which revealed increased expressions of NOS and hypoxia inducible factor α(HIF-1α). In conclusion, we demonstrated that intracardiac NO/NOS enhances goldfish heart performance, remarkably expanding its hypoxic tolerance.


Subject(s)
Hemodynamics , Hypoxia/physiopathology , Nitric Oxide/physiology , Animals , Goldfish
7.
Nitric Oxide ; 32: 1-12, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23545405

ABSTRACT

African lungfishes (Protopterus spp.) are obligate air breathers which enter in a prolonged torpor (aestivation) in association with metabolic depression, and biochemical and morpho-functional readjustments during the dry season. During aestivation, the lungfish heart continues to pump, while the skeletal muscle stops to function but can immediately contract during arousal. Currently, nothing is known regarding the orchestration of the multilevel rearrangements occurring in myotomal and myocardial muscles during aestivation and arousal. Because of its universal role in cardio-circulatory and muscle homeostasis, nitric oxide (NO) could be involved in coordinating these stress-induced adaptations. Western blotting and immunofluorescence microscopy on cardiac and skeletal muscles of Protopterus annectens (freshwater, 6months of aestivation and 6days after arousal) showed that expression, localization and activity of the endothelial-like nitric oxide synthase (eNOS) isoform and its partners Akt and Hsp-90 are tissue-specifically modulated. During aestivation, phospho-eNOS/eNOS and phospho-Akt/Akt ratios increased in the heart but decreased in the skeletal muscle. By contrast, Hsp-90 increased in both muscle types during aestivation. TUNEL assay revealed that increased apoptosis occurred in the skeletal muscle of aestivating lungfish, but the myocardial apoptotic rate of the aestivating lungfish remained unchanged as compared with the freshwater control. Consistent with the preserved cardiac activity during aestivation, the expression of apoptosis repressor (ARC) also remained unchanged in the heart of aestivating and aroused fish as compared with the freshwater control. Contrarily, ARC expression was strongly reduced in the skeletal muscle of aestivating lungfish. On the whole, our data indicate that changes in the eNOS/NO system and cell turnover are implicated in the morpho-functional readjustments occurring in lungfish cardiac and skeletal muscle during the switch from freshwater to aestivation, and between the maintenance and arousal phases of aestivation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Fishes/metabolism , Muscle, Skeletal/enzymology , Myocardium/enzymology , Nitric Oxide Synthase Type III/metabolism , Animals , Apoptosis/physiology , Estivation , Fresh Water , HSP90 Heat-Shock Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myocardium/cytology , Myocardium/metabolism , Proto-Oncogene Proteins c-akt/metabolism
8.
Article in English | MEDLINE | ID: mdl-23123761

ABSTRACT

The Frank-Starling law is a fundamental property of the vertebrate myocardium which allows, when the end-diastolic volume increases, that the consequent stretch of the myocardial fibers generates a more forceful contraction. It has been shown that in the eel (Anguilla anguilla) heart, nitric oxide (NO) exerts a direct myocardial relaxant effect, increasing the sensitivity of the Frank-Starling response (Garofalo et al., 2009). With the use of isolated working heart preparations, this study investigated the relationship between NO modulation of Frank-Starling response and temperature challenges in the eel. The results showed that while, in long-term acclimated fish (spring animals perfused at 20 °C and winter animals perfused at 10 °C) the inhibition of NO production by L-N5 (1-iminoethyl)ornithine (L-NIO) significantly reduced the Frank-Starling response, under thermal shock conditions (spring animals perfused at 10 or 15 °C and winter animals perfused at 15 or 20 °C) L-NIO treatment resulted without effect. Western blotting analysis revealed a decrease of peNOS and pAkt expressions in samples subjected to thermal shock. Moreover, an increase in Hsp90 protein levels was observed under heat thermal stress. Together, these data suggest that the NO synthase/NO-dependent modulation of the Frank-Starling mechanism in fish is sensitive to thermal stress.


Subject(s)
Acclimatization/radiation effects , Eels/physiology , Heart , Nitric Oxide , Acclimatization/physiology , Animals , HSP90 Heat-Shock Proteins/metabolism , Heart/drug effects , Heart/physiology , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Nitric Oxide/metabolism , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Organ Culture Techniques , Ornithine/analogs & derivatives , Ornithine/pharmacology , Temperature
10.
Curr Med Chem ; 19(24): 4086-92, 2012.
Article in English | MEDLINE | ID: mdl-22834799

ABSTRACT

Serpinins are a family of peptides derived from proteolytic cleavage of the penultimate and the last pair of basic residues at the C-terminus of Chromogranin A. Three forms of naturally occurring serpinin have been found in AtT-20 pituitary cells and rat heart. They are serpinin, pyrogutaminated (pGlu) -serpinin and a C-terminally extended form, serpinin-RRG. In addition pGlu-serpinin has been found in brain, primarily in neurites and nerve terminals and shown to have protective effects against oxidative stress on neurons and pituitary cells. Serpinin has also been demonstrated to regulate granule biogenesis in endocrine cells by up-regulating the protease inhibitor, protease nexin-1 transcription via a cAMP-PKA-sp1 pathway. This leads to inhibition of granule protein degradation in the Golgi complex which in turn promotes granule formation. More recently, pGlu-serpinin has been demonstrated to enhance both myocardial contractility (inotropy) and relaxation (lusitropy). In the Langendorff perfused rat heart, pGlu-serpinin showed a concentration-dependent positive inotropic effect exerted through a cAMP-PKA dependent pathway. In conclusion, the serpinin peptides have profound effects at many levels that affect the endocrine and nervous systems and cardiac function.


Subject(s)
Cell Death/drug effects , Myocardial Contraction/drug effects , Serpins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromogranin A/chemistry , Chromogranin A/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Golgi Apparatus/metabolism , Molecular Sequence Data , Myocardium/metabolism , Neurons/metabolism , Rats , Serpin E2/genetics , Serpin E2/metabolism , Serpins/chemistry , Serpins/pharmacology , Signal Transduction , Up-Regulation/drug effects
11.
Article in English | MEDLINE | ID: mdl-22705557

ABSTRACT

Using morphological and physiological approaches we provided, for the first time, a structural and functional characterization of Carassius auratus L. heart. Besides to the classical four chambers, i.e. sinus venosus, atrium, ventricle, bulbus, we described two distinct structures corresponding to the atrio-ventricular (AV) region and the conus arteriosus. The atrium is very large and highly trabeculated; the ventricle shows an outer compacta, vascularized by coronary vessels, and an inner spongiosa; the bulbus wall is characterized by a high elastin/collagen ratio, which makes it extremely compliant. Immunolocalization revealed a strong expression of activated "eNOS-like" isoforms both at coronary endothelium and, to a lesser extent, in the myocardiocytes and the endocardial endothelium (EE). The structural design of the heart appears to comply with its mechanical function. Using an in vitro working heart preparation, cardiac performance was evaluated at different filling and afterload pressures. The hearts were very sensitive to filling pressure increases. Maximum Stroke volume (SV=1.08 ± 0.09 mL/kg body mass) was obtained with an input pressure of 0.4 kPa. The heart was not able to sustain afterload increases, values higher than 1.5 kPa impairing its performance. These morpho-functional features are consistent with a volume pump mechanical performance.


Subject(s)
Goldfish/anatomy & histology , Heart/anatomy & histology , Animals , Coronary Vessels/anatomy & histology , Fish Proteins/metabolism , Goldfish/physiology , Heart Rate , In Vitro Techniques , Myocardium/enzymology , Nitric Oxide Synthase Type III/metabolism , Stroke Volume
12.
Nutr Metab Cardiovasc Dis ; 21(5): 362-71, 2011 May.
Article in English | MEDLINE | ID: mdl-20096547

ABSTRACT

BACKGROUND AND AIMS: Moderate red wine consumption associates with lower incidence of cardiovascular diseases. Attention to the source of this cardioprotection was focused on flavonoids, the non-alcoholic component of the red wine, whose intake inversely correlates with adverse cardiovascular events. We analysed whether two red wine flavonoids, quercetin and myricetin, affect mammalian basal myocardial and coronary function. METHODS AND RESULTS: Quercetin and myricetin effects were evaluated on isolated and Langendorff perfused rat hearts under both basal conditions and α- and ß-adrenergic stimulation. The intracellular signalling involved in the effects of these flavonoids was analysed on perfused hearts and by western blotting on cardiac and HUVEC extracts. Quercetin induced biphasic inotropic and lusitropic effects, positive at lower concentrations and negative at higher concentrations. Contrarily, Myricetin elicits coronary dilation, without affecting contractility and relaxation. Simultaneous administration of the two flavonoids only induced vasodilation. Quercetin-elicited positive inotropism and lusitropism depend on ß1/ß2-adrenergic receptors and associate with increased intracellular cAMP, while the negative inotropism and lusitropism observed at higher concentrations were α-adrenergic-dependent. NOS inhibition abolished Myricetin-elicited vasodilation, also inducing Akt, ERK1/2 and eNOS phosphorylation in both ventricles and HUVEC. Myricetin-dependent vasodilation increases intracellular cGMP and is abolished by triton X-100. CONCLUSIONS: The cardiomodulation elicited on basal mechanical performance by quercetin and the selective vasodilation induced by myricetin point to these flavonoids as potent cardioactive principles, able to protect the heart in the presence of cardiovascular diseases.


Subject(s)
Antioxidants/pharmacology , Flavonoids/pharmacology , Heart/drug effects , Myocardium/metabolism , Quercetin/pharmacology , Signal Transduction , Wine , Analysis of Variance , Animals , In Vitro Techniques , Male , Octoxynol , Rats , Rats, Wistar , Vasodilation/drug effects
13.
Curr Med Chem ; 17(18): 1915-25, 2010.
Article in English | MEDLINE | ID: mdl-20377513

ABSTRACT

Recently, the circulating anion nitrite (NO2-), the largest physiological reservoir of nitric oxide (NO) in the body, has revealed itself as a signalling molecule mediating numerous biological responses. Since it was estimated that as much as 70% of plasma nitrite originates from nitric oxide synthases (NOSs), mainly in the endothelium by endothelial NOS, nitrite is considered an index of NOSs activity. Exogenous sources, principally environmental pollutants and intake of vegetables, also contribute to this NO reserve. In mammalian blood, nitrite, present at nanomolar concentrations, can be reduced to bioactive NO along a physiological oxygen and pH gradient either non-enzymatically (acidic disproportionation) or by a number of enzymes including xanthine oxidoreductase, NOS, mitochondrial cytochromes and deoxygenated haemoglobin and myoglobin. The various NO-dependent nitrite-induced biological responses include hypoxic vasodilation, inhibition of mitochondrial respiration, cytoprotection following ischemia/reperfusion, and regulation of protein and gene expression. Since NO is a major paracrine-autocrine cardiovascular modulator and nitrite acts mainly as an endocrine store of NO, it is not surprising that NO2 - exerts important cardiovascular actions both under normal and physio-pathological conditions. In the interdisciplinary framework of the NO cycle concept, this review illustrates the actions exerted by nitrite on the cardiovascular system. Since the majority of the NO2 - -oriented studies focused on the systemic and regional control of blood flow both under physiological and ischemia/reperfusion conditions, we will firstly consider this issue. Secondly, the nitrite- induced effects on myocardial contractile and relaxation processes will be discussed, emphasizing the biomedical interest of nitrite as a new therapeutic agent. The importance of cardiac myoglobin as nitrite-reductase able to exert cardioprotection through a novel function, in addition to its role as classical respiratory protein, will be highlighted. Finally, using recent data from others and our labs, we will emphasize the importance of fish and amphibian heart models with diverse morphologies and blood supply for providing remarkable insights on "ancestral" functions of the nitrite-NO system in vertebrates, which, in turn, may help to expand its actual significance in human physiology.


Subject(s)
Cardiotonic Agents/metabolism , Cardiotonic Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular System/metabolism , Nitrites/metabolism , Nitrites/therapeutic use , Animals , Cardiovascular System/pathology , Humans , Nitric Oxide/metabolism , Nitrite Reductases/metabolism
14.
Proc Biol Sci ; 276(1675): 4043-52, 2009 Nov 22.
Article in English | MEDLINE | ID: mdl-19726482

ABSTRACT

The Frank-Starling mechanism is a fundamental property of the vertebrate heart, which allows the myocardium to respond to increased filling pressure with a more vigorous contraction of its lengthened fibres. In mammals, myocardial stretch increases cardiac nitric oxide (NO) release from both vascular endothelium and cardiomyocytes. This facilitates myocardial relaxation and ventricular diastolic distensibility, thus influencing the Frank-Starling mechanism. In the in vitro working heart of the eel Anguilla anguilla, we previously showed that an endogenous NO release affects the Frank-Starling response making the heart more sensitive to preload. Using the same bioassay, we now demonstrate that this effect is confirmed in the presence of the exogenous NO donor S-nitroso-N-acetyl penicillamine, is independent from endocardial endothelium and guanylate cyclase/cGMP/protein kinase G and cAMP/protein kinase A pathways, involves a PI(3)kinase-mediated activation of endothelial NO synthase and a modulation of the SR-CA(2+)ATPase (SERCA2a) pumps. Furthermore, we show that NO influences cardiac response to preload through S-nitrosylation of phospholamban and consequent activation of SERCA2a. This suggests that in the fish heart NO modulates the Frank-Starling response through a beat-to-beat regulation of calcium reuptake and thus of myocardial relaxation. We propose that this mechanism represents an important evolutionary step for the stretch-induced intrinsic regulation of the vertebrate heart, providing, at the same time, a stimulus for mammalian-oriented studies.


Subject(s)
Calcium-Binding Proteins/pharmacology , Eels/physiology , Heart/physiology , Animals , Calcium-Binding Proteins/metabolism , Cardiac Output/drug effects , Coronary Circulation/drug effects , Cyclic AMP/metabolism , Endocardium/physiology , Endothelium/metabolism , Gene Expression Regulation/physiology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , S-Nitroso-N-Acetylpenicillamine/pharmacology , Signal Transduction , Ventricular Pressure/physiology
15.
Article in English | MEDLINE | ID: mdl-19401238

ABSTRACT

The teleostean Channichthyidae (icefish), endemic stenotherms of the Antarctic waters, perennially at or near freezing, represent a unique example of disaptation among adult vertebrates for their loss of functional traits, particularly hemoglobin (Hb) and, in some species, cardiac myoglobin (Mb), once considered to be essential-life oxygen-binding chromoproteins. Conceivably, this stably frigid, oxygen-rich habitat has permitted high tolerance of disaptation, followed by subsequent adaptive recovery based on gene expression reprogramming and compensatory responses, including an alternative cardio-circulatory design, Hb-free blood and Mb-free cardiac muscle. This review revisits the functional significance of the multilevel cardio-circulatory compensations (hypervolemia, near-zero hematocrit and low blood viscosity, large bore capillaries, increased vascularity with great capacitance, cardiomegaly with very large cardiac output, high blood flow with low systemic pressure and systemic resistance) that counteract the challenge of hypoxemic hypoxia by increasing peripheral oxygen transcellular movement for aerobic tissues, including the myocardium. Reconsidered in the context of recent knowledge on both polar cold adaptation and the new questions related to the advent of nitric oxide (NO) biology, these compensations can be interpreted either according to the "loss-without-penalty" alternative, or in the context of an excessive environmental oxygen supply at low cellular cost and oxygen requirement in the cold. Therefore, rather than reflecting oxygen limitation, several traits may indicate structural overcompensation of oxygen supply reductions at cell/tissue levels. At the multilevel cardio-circulatory adjustments, NO is revealing itself as a major integrator, compensating disaptation with functional phenotypic plasticity, as illustrated by the heart paradigm. Beside NOS-dependent NO generation, recent knowledge concerning Hb/Mb interplay with NO and nitrite has revealed unexpected functions in addition to the classical respiratory role of these proteins. In fact, nitrite, a major biologic reservoir of NO, generates it through deohyHb- and deoxyMb-dependent nitrite reduction, thereby regulating hypoxic vasodilation, cellular respiration and signalling. We suggest that both Hb and Mb are involved as nitrite reductases under hypoxic conditions in a number of cardiocirculatory processes. On the whole, this opens new horizons in environmental and evolutionary physiology.


Subject(s)
Adaptation, Physiological/physiology , Perciformes/physiology , Acclimatization , Animals , Biological Evolution , Cardiovascular Physiological Phenomena , Cold Temperature , Heart/anatomy & histology , Heart/physiology , Hemoglobins/genetics , Homeostasis/physiology , Mitochondria, Heart , Myocardium/metabolism , Myoglobin/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase/physiology , Oxygen/blood , Phenotype
16.
Biochim Biophys Acta ; 1787(7): 849-55, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19298789

ABSTRACT

Being the largest form of intravascular and tissue storage of nitric oxide (NO) and a signalling molecule itself, the nitrite anion (NO(2)(-)) has emerged as a key player in many biological processes. Since the heart is under an important NO-mediated autocrine-paracrine control, in mammals the cardiac effects of nitrite are under intensive investigation. In contrast, nothing is known in non-mammalian vertebrates. We evaluated nitrite influence on cardiac performance in the perfused beating heart of three different cold-blooded vertebrates, i.e. two teleost fishes, the temperate red-blooded Anguilla anguilla, the Antarctic stenotherm, hemoglobinless Chionodraco hamatus (icefish), and the frog Rana esculenta. We showed that, under basal conditions, in all animals nitrite influences cardiac mechanical performance, inducing negative inotropism in eel and frog, while being a positive inotrope in C. hamatus. In all species, these responses parallel the inotropic effects of authentic NO. We also demonstrated that the nitrite-dependent inotropic effects are i) dependent from NO synthase (NOS) activity in fish; ii) sensitive to NO scavenging in frog; iii) cGMP/PKG-dependent in both eel and frog. Results suggest that nitrite is an integral physiological source of NO and acts as a signalling molecule in lower vertebrate hearts, exerting relevant inotropic actions through different species-specific mechanisms.


Subject(s)
Anguilla/metabolism , Myocardial Contraction/drug effects , Nitrites/pharmacology , Perciformes/metabolism , Rana esculenta/metabolism , Animals , Antarctic Regions , Female , Male , Myocardium/metabolism , Oceans and Seas
17.
FASEB J ; 22(11): 3992-4004, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18697842

ABSTRACT

Chromogranin A (CGA), produced by human and rat myocardium, generates several biologically active peptides processed at specific proteolytic cleavage sites. A highly conserved cleavage N-terminal site is the bond 64-65 that reproduces the native rat CGA sequence (rCGA1-64), corresponding to human N-terminal CGA-derived vasostatin-1. rCGA1-64 cardiotropic activity has been explored in rat cardiac preparations. In Langendorff perfused rat heart, rCGA1-64 (from 33 nM) induced negative inotropism and lusitropism as well as coronary dilation, counteracting isoproterenol (Iso) - and endothelin-1 (ET-1) -induced positive inotropic effects and ET-1-dependent coronary constriction. rCGA1-64 also depressed basal and Iso-induced contractility on rat papillary muscles, without affecting calcium transients on isolated ventricular cells. Structure-function analysis using three modified peptides on both rat heart and papillary muscles revealed the disulfide bridge requirement for the cardiotropic action. A decline in Iso intrinsic activity in the presence of the peptides indicates a noncompetitive antagonistic action. Experiments on rat isolated cardiomyocytes and bovine aortic endothelial cells indicate that the negative inotropism observed in rat papillary muscle is probably due to an endothelial phosphatidylinositol 3-kinase-dependent nitric oxide release, rather than to a direct action on cardiomyocytes. Taken together, our data strongly suggest that in the rat heart the homologous rCGA1-64 fragment exerts an autocrine/paracrine modulation of myocardial and coronary performance acting as stabilizer against intense excitatory stimuli.


Subject(s)
Chromogranin A/metabolism , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Papillary Muscles/metabolism , Vasodilation/physiology , Animals , Aorta/cytology , Aorta/metabolism , Autocrine Communication/drug effects , Autocrine Communication/physiology , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cattle , Chromogranin A/pharmacology , Endothelial Cells/cytology , Endothelin-1/pharmacology , Humans , Isoproterenol/pharmacology , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Papillary Muscles/cytology , Paracrine Communication/drug effects , Paracrine Communication/physiology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar , Structure-Activity Relationship , Vasodilation/drug effects
18.
Curr Med Chem ; 15(14): 1444-51, 2008.
Article in English | MEDLINE | ID: mdl-18537621

ABSTRACT

Over the last 50 years, increasing evidence has documented the ability of cardiac non-neuronal cells to synthesize and release catecholamines (CAs) and the vasorelaxant natriuretic peptides (NPs), which both regulate cardiovascular homeostasis in health and disease. This knowledge has firmly established the concept of the heart as an endocrine organ. The contents of this frame have been richly expanded by the identification of an increasing number of intracardiac endocrine modulators, including Chromogranin-A (CgA) and its derived peptides. In the rat heart, CgA is co-stored and co-released with Atrial NP (ANP) in non-adrenergic myoendocrine atrial cells as well as in atrial and ventricular Purkinje fibres. In the ventricular myocardium of the human hypertrophic and dilated heart, CgA co-localizes with B-type NP (BNP). CgA is the precursor of biologically active peptides produced by proteolytic cleavage. One of them, the human recombinant 1-76 CgA-derived vasostatin-1 (VS-1), is an inhibitor of cardiac contraction and relaxation, a non-competitive counter-regulator of beta-adrenergic stimulation and a protecting agent in ischemic preconditioning. Therefore, it may function as a cardiocirculatory homeostatic stabilizer, particularly in the presence of intense adrenergic stimuli, e. g. under stress responses. Since in patients with chronic heart failure circulating CgA levels increase up to 10-20 nM, depending on the severity of the disease and are independent prognostic indicators of mortality, knowledge on the physio-pathological significance of locally produced and/or circulating CgA-derived peptides, as attemped in this synopsis, may pave the way for clinically-oriented cardiovascular applications.


Subject(s)
Chromogranin A/metabolism , Heart/drug effects , Myocardium/metabolism , Peptide Fragments/pharmacology , Animals , Chromogranin A/pharmacology , Cytoskeleton/metabolism , Heart/physiology , Hormones/metabolism , Humans , Myocardial Contraction/drug effects , Natriuretic Peptide, Brain/metabolism , Neurosecretory Systems/physiology , Peptide Fragments/metabolism , Sympathetic Nervous System/physiology
19.
Cell Mol Life Sci ; 64(22): 2863-86, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17717629

ABSTRACT

Chromogranin A (CgA) belongs to the granin family of uniquely acidic secretory proteins co-stored and co-secreted with other hormones and peptides in elements of the diffuse neuroendocrine system. The granins arise from different genes and are characterized by numerous sites for post-translational cleavage into shorter peptides with postulated regulatory properties. This review is directed towards endocrine aspects of CgA and its biologically active peptides. There is ample evidence from in vitro studies of distinct effects and targets for three CgA-derived peptides, vasostatin-I, pancreastatin and catestatin. Endocrine regulations are indicated from in vivo studies, consistent with the postulated prohormone function of CgA for peptides with regulatory properties. Most of the effects fit into patterns of direct or indirect, inhibitory modulations of major functions, implicating CgA peptides in regulation of calcium and glucose metabolism, cardiovascular functions, gastrointestinal motility and nociception, tissue repair, inflammatory responses and as host defense peptides in the first phase of microbial invasions.


Subject(s)
Chromogranin A/physiology , Endocrine Glands/physiology , Animals , Calcium/metabolism , Carbohydrate Metabolism , Cardiovascular Physiological Phenomena , Chromogranin A/chemistry , Humans , Immunity, Innate , Inflammation/physiopathology , Models, Biological , Models, Molecular , Neoplasms/pathology , Neoplasms/physiopathology , Neuroendocrine Tumors/physiopathology , Neurosecretory Systems/physiology , Pancreatic Hormones/chemistry , Pancreatic Hormones/physiology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Structure-Activity Relationship
20.
J Exp Biol ; 209(Pt 24): 4966-73, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17142685

ABSTRACT

Neuroendocrine regulation of cardiac function involves a population of three types of beta-adrenoceptors (ARs). In various mammalian species, beta1- and beta2-AR stimulation produces an increase in contractility; whereas beta3-AR activation mediates negative inotropic effects. At the moment, nothing is known about the physiological role of beta3-AR in fish. Using an isolated working heart preparation, we show that a beta3-AR selective agonist BRL(37344) (0.1-100 nmol l(-1)) elicits a dose-dependent negative inotropism in the freshwater eel Anguilla anguilla. This effect was insensitive to the beta1/beta2-AR inhibitor nadolol (10 mumol l(-1)), but was blocked by the beta3-AR-specific antagonist SR(59230) (10 nmol l(-1)). The analysis of the percentage of stroke work (SW) variations, in terms of EC(50) values, induced by BRL(37344) alone (10 nmol l(-1)), and in presence of SR(59230) (10 nmol l(-1)), indicated a competitive antagonism of SR(59230). In addition to the classic positive inotropism, the non-specific beta agonist isoproterenol (100 nmol l(-1)) induced, in 30% of the preparations, a negative inotropic effect that was abrogated by pre-treatment with SR(59230), pointing to a beta3-mediated pathway. The BRL(37344)-induced negative inotropic effect was abolished by exposure to a G(i/o) proteins inhibitor pertussis toxin (PTx; 0.01 nmol l(-1)), suggesting a G(i/o)-dependent mechanism. Using L-N5(l-imino-ethyl)ornithine (L-NIO; 10 mumol l(-1)), as a nitric oxide (NO) synthase (NOS) blocker and haemoglobin (Hb; 1 mumol l(-1)), as a NO scavenger, we demonstrated that NO signalling is involved in the BRL(37344)-induced response. Pre-treatment with either an inhibitor of soluble guanylate cyclase (GC) 1H-(1,2,4) oxadiazolo-(4,3-a)quinoxalin-1-one (ODQ; 10 mumol l(-1)), or an inhibitor of the cGMP-activated protein kinase (PKG) KT(5823) (100 nmol l(-1)), abolished the beta3-dependent negative inotropism, indicating the cGMP-PKG component as a crucial target of NO signalling. Taken together, our findings provide functional evidence for the presence of beta3-like adrenoceptors in the eel Anguilla anguilla heart identifying, for the first time in a working fish heart, the beta3-AR-dependent negative inotropy discovered in mammals.


Subject(s)
Anguilla/physiology , Cyclic GMP/metabolism , Myocardial Contraction/physiology , Nitric Oxide/metabolism , Receptors, Adrenergic, beta-3/physiology , Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cyclic GMP-Dependent Protein Kinases/metabolism , Depression, Chemical , Ethanolamines/pharmacology , GTP-Binding Proteins/metabolism , Heart/drug effects , Heart/physiology , In Vitro Techniques , Isoproterenol/pharmacology , Myocardial Contraction/drug effects , Nadolol/pharmacology , Propanolamines/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...