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










Publication year range
1.
Circ Res ; 134(8): 990-1005, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38456287

ABSTRACT

BACKGROUND: Growing evidence correlated changes in bioactive sphingolipids, particularly S1P (sphingosine-1-phosphate) and ceramides, with coronary artery diseases. Furthermore, specific plasma ceramide species can predict major cardiovascular events. Dysfunction of the endothelium lining lesion-prone areas plays a pivotal role in atherosclerosis. Yet, how sphingolipid metabolism and signaling change and contribute to endothelial dysfunction and atherosclerosis remain poorly understood. METHODS: We used an established model of coronary atherosclerosis in mice, combined with sphingolipidomics, RNA-sequencing, flow cytometry, and immunostaining to investigate the contribution of sphingolipid metabolism and signaling to endothelial cell (EC) activation and dysfunction. RESULTS: We demonstrated that hemodynamic stress induced an early metabolic rewiring towards endothelial sphingolipid de novo biosynthesis, favoring S1P signaling over ceramides as a protective response. This finding is a paradigm shift from the current belief that ceramide accrual contributes to endothelial dysfunction. The enzyme SPT (serine palmitoyltransferase) commences de novo biosynthesis of sphingolipids and is inhibited by NOGO-B (reticulon-4B), an ER membrane protein. Here, we showed that NOGO-B is upregulated by hemodynamic stress in myocardial EC of ApoE-/- mice and is expressed in the endothelium lining coronary lesions in mice and humans. We demonstrated that mice lacking NOGO-B specifically in EC (Nogo-A/BECKOApoE-/-) were resistant to coronary atherosclerosis development and progression, and mortality. Fibrous cap thickness was significantly increased in Nogo-A/BECKOApoE-/- mice and correlated with reduced necrotic core and macrophage infiltration. Mechanistically, the deletion of NOGO-B in EC sustained the rewiring of sphingolipid metabolism towards S1P, imparting an atheroprotective endothelial transcriptional signature. CONCLUSIONS: These data demonstrated that hemodynamic stress induced a protective rewiring of sphingolipid metabolism, favoring S1P over ceramide. NOGO-B deletion sustained the rewiring of sphingolipid metabolism toward S1P protecting EC from activation under hemodynamic stress and refraining coronary atherosclerosis. These findings also set forth the foundation for sphingolipid-based therapeutics to limit atheroprogression.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Humans , Animals , Mice , Ceramides/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Nogo Proteins , Sphingolipids/metabolism , Sphingosine/metabolism , Lysophospholipids/metabolism , Endothelium/metabolism , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Apolipoproteins E
2.
Cardiovasc Res ; 119(2): 506-519, 2023 03 31.
Article in English | MEDLINE | ID: mdl-35815623

ABSTRACT

AIMS: Growing evidence correlate the accrual of the sphingolipid ceramide in plasma and cardiac tissue with heart failure (HF). Regulation of sphingolipid metabolism in the heart and the pathological impact of its derangement remain poorly understood. Recently, we discovered that Nogo-B, a membrane protein of endoplasmic reticulum, abundant in the vascular wall, down-regulates the sphingolipid de novo biosynthesis via serine palmitoyltransferase (SPT), first and rate liming enzyme, to impact vascular functions and blood pressure. Nogo-A, a splice isoform of Nogo, is transiently expressed in cardiomyocyte (CM) following pressure overload. Cardiac Nogo is up-regulated in dilated and ischaemic cardiomyopathies in animals and humans. However, its biological function in the heart remains unknown. METHODS AND RESULTS: We discovered that Nogo-A is a negative regulator of SPT activity and refrains ceramide de novo biosynthesis in CM exposed to haemodynamic stress, hence limiting ceramide accrual. At 7 days following transverse aortic constriction (TAC), SPT activity was significantly up-regulated in CM lacking Nogo-A and correlated with ceramide accrual, particularly very long-chain ceramides, which are the most abundant in CM, resulting in the suppression of 'beneficial' autophagy. At 3 months post-TAC, mice lacking Nogo-A in CM showed worse pathological cardiac hypertrophy and dysfunction, with ca. 50% mortality rate. CONCLUSION: Mechanistically, Nogo-A refrains ceramides from accrual, therefore preserves the 'beneficial' autophagy, mitochondrial function, and metabolic gene expression, limiting the progression to HF under sustained stress.


Subject(s)
Heart Failure , Sphingolipids , Humans , Mice , Animals , Nogo Proteins/genetics , Nogo Proteins/metabolism , Sphingolipids/metabolism , Ceramides/metabolism , Heart Failure/genetics , Myocytes, Cardiac/metabolism
3.
Nat Commun ; 13(1): 6207, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266273

ABSTRACT

Biological tissues comprise a spatially complex structure, composition and organization at the microscale, named the microstructure. Given the close structure-function relationships in tissues, structural characterization is essential to fully understand the functioning of healthy and pathological tissues, as well as the impact of possible treatments. Here, we present a nondestructive imaging approach to perform quantitative 3D histo(patho)logy of biological tissues, termed Cryogenic Contrast-Enhanced MicroCT (cryo-CECT). By combining sample staining, using an X-ray contrast-enhancing staining agent, with freezing the sample at the optimal freezing rate, cryo-CECT enables 3D visualization and structural analysis of individual tissue constituents, such as muscle and collagen fibers. We applied cryo-CECT on murine hearts subjected to pressure overload following transverse aortic constriction surgery. Cryo-CECT allowed to analyze, in an unprecedented manner, the orientation and diameter of the individual muscle fibers in the entire heart, as well as the 3D localization of fibrotic regions within the myocardial layers. We foresee further applications of cryo-CECT in the optimization of tissue/food preservation and donor banking, showing that cryo-CECT also has clinical and industrial potential.


Subject(s)
Musculoskeletal System , Mice , Animals , X-Ray Microtomography/methods , Freezing , Staining and Labeling , Collagen , Imaging, Three-Dimensional/methods
5.
Free Radic Biol Med ; 166: 238-254, 2021 04.
Article in English | MEDLINE | ID: mdl-33675956

ABSTRACT

Heart failure is one of the leading causes of death and disability worldwide. Left ventricle remodeling, fibrosis, and ischemia/reperfusion injury all contribute to the deterioration of cardiac function and predispose to the onset of heart failure. Adenosine monophosphate-activated protein kinase (AMPK) is the universally recognized energy sensor which responds to low ATP levels and restores cellular metabolism. AMPK activation controls numerous cellular processes and, in the heart, it plays a pivotal role in preventing onset and progression of disease. Excessive reactive oxygen species (ROS) generation, known as oxidative stress, can activate AMPK, conferring an additional role of AMPK as a redox-sensor. In this review, we discuss recent insights into the crosstalk between ROS and AMPK. We describe the molecular mechanisms by which ROS activate AMPK and how AMPK signaling can further prevent heart failure progression. Ultimately, we review the potential therapeutic approaches to target AMPK for the treatment of cardiovascular disease and prevention of heart failure.


Subject(s)
AMP-Activated Protein Kinases , Myocardium , AMP-Activated Protein Kinases/genetics , Adenosine Monophosphate , Myocytes, Cardiac , Reactive Oxygen Species
6.
Am J Physiol Heart Circ Physiol ; 320(2): H838-H853, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33416451

ABSTRACT

Although sodium glucose cotransporter 1 (SGLT1) has been identified as one of the major SGLT isoforms expressed in the heart, its exact role remains elusive. Evidence using phlorizin, the most common inhibitor of SGLTs, has suggested its role in glucose transport. However, phlorizin could also affect classical facilitated diffusion via glucose transporters (GLUTs), bringing into question the relevance of SGLT1 in overall cardiac glucose uptake. Accordingly, we assessed the contribution of SGLT1 in cardiac glucose uptake using the SGLT1 knockout mouse model, which lacks exon 1. Glucose uptake was similar in cardiomyocytes isolated from SGLT1-knockout (Δex1KO) and control littermate (WT) mice either under basal state, insulin, or hyperglycemia. Similarly, in vivo basal and insulin-stimulated cardiac glucose transport measured by micro-PET scan technology did not differ between WT and Δex1KO mice. Micromolar concentrations of phlorizin had no impact on glucose uptake in either isolated WT or Δex1KO-derived cardiomyocytes. However, higher concentrations (1 mM) completely inhibited insulin-stimulated glucose transport without affecting insulin signaling nor GLUT4 translocation independently from cardiomyocyte genotype. Interestingly, we discovered that mouse and human hearts expressed a shorter slc5a1 transcript, leading to SGLT1 protein lacking transmembrane domains and residues involved in glucose and sodium bindings. In conclusion, cardiac SGLT1 does not contribute to overall glucose uptake, probably due to the expression of slc5a1 transcript variant. The inhibitory effect of phlorizin on cardiac glucose uptake is SGLT1-independent and can be explained by GLUT transporter inhibition. These data open new perspectives in understanding the role of SGLT1 in the heart.NEW & NOTEWORTHY Ever since the discovery of its expression in the heart, SGLT1 has been considered as similar as the intestine and a potential contributor to cardiac glucose transport. For the first time, we have demonstrated that a slc5a1 transcript variant is present in the heart that has no significant impact on cardiac glucose handling.


Subject(s)
Glucose/metabolism , Myocytes, Cardiac/metabolism , Sodium-Glucose Transporter 1/metabolism , Animals , Biological Transport , Cells, Cultured , Glucose Transporter Type 4/antagonists & inhibitors , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Phlorhizin/pharmacology , Protein Isoforms , Rats, Wistar , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 1/genetics
7.
JCI Insight ; 4(9)2019 05 02.
Article in English | MEDLINE | ID: mdl-31045580

ABSTRACT

Hypercholesterolemia and hypertension are two major risk factors for coronary artery diseases, which remain the major cause of mortality in the industrialized world. Current animal models of atherosclerosis do not recapitulate coronary plaque disruption, thrombosis, and myocardial infarction occurring in humans. Recently, we demonstrated that exposure of the heart to high pressure, by transverse aortic constriction (TAC), induced coronary lesions in ApoE-/- mice on chow diet. The aim of this study was to characterize the magnitude and location of coronary lesions in ApoE-/- mice after TAC and to assess the susceptibility of coronary plaque to disruption, leading to myocardial events. Here, we describe a reliable pathological condition in mice characterized by the development of coronary lesions and its progression, leading to myocardial infarction; this model better recapitulates human disease. Following TAC surgery, about 90% of ApoE-/- mice developed coronary lesions, especially in the left anterior descending artery, with 59% of the mice manifesting a different magnitude of LAD stenosis. Myocardial events, identified in 74% of the mice, were mainly due to coronary plaque thrombosis and occlusion. That TAC-induced development and progression of coronary lesions in ApoE-/- mice, leading to myocardial events, represents a potentially novel and important tool to investigate the development of coronary lesions and its sequelae in a setting that better resemble human conditions.


Subject(s)
Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/pathology , Disease Progression , Heart/physiopathology , Myocardium/pathology , Plaque, Atherosclerotic/pathology , Animals , Coronary Artery Disease/complications , Coronary Vessels/pathology , Disease Models, Animal , Embolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology
8.
J Pharmacol Exp Ther ; 366(2): 314-321, 2018 08.
Article in English | MEDLINE | ID: mdl-29907698

ABSTRACT

We previously discovered that oral treatment with AC261066, a synthetic selective agonist for the retinoic acid ß2-receptor, decreases oxidative stress in the liver, pancreas, and kidney of mice fed a high-fat diet (HFD). Since hyperlipidemic states are causally associated with myocardial ischemia and oxidative stress, we have now investigated the effects of AC261066 in an ex vivo ischemia/reperfusion (I/R) injury model in hearts of two prototypic dysmetabolic mice. We found that a 6-week oral treatment with AC261066 in both genetically hypercholesterolemic (ApoE-/-) and obese (HFD-fed) wild-type mice exerts protective effects when their hearts are subsequently subjected to I/R ex vivo in the absence of added drug. In ApoE-/- mice this cardioprotection ensued without hyperlipidemic changes. Cardioprotection consisted of attenuation of infarct size, diminution of norepinephrine (NE) spillover, and alleviation of reperfusion arrhythmias. This cardioprotection was associated with a reduction in oxidative stress and mast cell (MC) degranulation. We suggest that the reduction in myocardial injury and adrenergic activation, and the antiarrhythmic effects, result from decreased formation of oxygen radicals and toxic aldehydes known to elicit the release of MC-derived renin, promoting the activation of the local renin-angiotensin system leading to enhanced NE release and reperfusion arrhythmias. Because these beneficial effects of AC261066 occurred at the ex vivo level following oral drug treatment, our data suggest that AC261066 could be viewed as a therapeutic means to reduce I/R injury of the heart, and potentially also be considered in the treatment of other cardiovascular ailments such as chronic arrhythmias and cardiac failure.


Subject(s)
Benzoates/pharmacology , Cardiotonic Agents/pharmacology , Receptors, Retinoic Acid/agonists , Thiazoles/pharmacology , Animals , Mast Cells/drug effects , Mast Cells/immunology , Mice , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress/drug effects
9.
Curr Med Chem ; 25(34): 4416-4431, 2018.
Article in English | MEDLINE | ID: mdl-29446730

ABSTRACT

BACKGROUND: Excessive norepinephrine (NE) release in the ischemic heart elicits severe and often lethal arrhythmias. Resident cardiac mast cells synthesize and store active renin, which is released upon degranulation, causing the activation of a local cardiac renin-angiotensin system (RAS) responsible for NE release and consequent arrhythmias. Toxic aldehydes, known to be formed by lipid peroxidation in ischemia/reperfusion (I/R), have been shown to degranulate mast cells and activate a local RAS. OBJECTIVE: To provide an up-to-date description of the roles of ischemic preconditioning (IPC) and Gicoupled receptors in anti-RAS cardioprotection. METHODS: Ex-vivo I/R models in cavian and murine hearts, and human and murine mast cell lines in vitro. RESULTS: IPC not only drastically reduces the injury subsequent to a prolonged ischemic event, but also decreases mast cell renin release, thus affording anti-RAS cardioprotection. Similarly, activation of Gicoupled receptors, such as histamine-H4, adenosine-A3 and sphingosine-1-phosphate-S1P1 receptors, all expressed at the mast cell surface, mimic the cardioprotective anti-RAS effects of IPC. The mechanism of this action depends on the sequential activation of a specific isoform of protein kinase C, PKCε, and mitochondrial aldehyde dehydrogenase-type 2 (ALDH2). Increased ALDH2 enzymatic activity exerts a pivotal role in the sequential inhibition of aldehyde-induced mast-cell renin release, prevention of RAS activation, reduction of NE release and alleviation of reperfusion arrhythmias. CONCLUSION: These recently discovered protective pathways indicate that activation of mast-cell Gicoupled receptors and subsequent ALDH2 phosphorylation/activation represent a novel therapeutic target for the alleviation of RAS-induced cardiac dysfunctions, including ischemic heart disease and congestive heart failure.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Myocardial Ischemia/pathology , Protein Kinase C-epsilon/metabolism , Animals , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Humans , Mast Cells/cytology , Mast Cells/drug effects , Mast Cells/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/prevention & control , Norepinephrine/metabolism
10.
J Pharmacol Exp Ther ; 362(2): 230-242, 2017 08.
Article in English | MEDLINE | ID: mdl-28500264

ABSTRACT

In the ischemic-reperfused (I/R) heart, renin-containing mast cells (MC) release enzymatically active renin, activating a local renin-angiotensin system (RAS), causing excessive norepinephrine release and arrhythmic dysfunction. Activation of Gi-receptors on MC and/or ischemic preconditioning (IPC) prevent renin release, thus providing anti-RAS cardioprotection. We questioned whether sphingosine-1-phosphate (S1P), a sphingolipid produced in the I/R heart, might afford anti-RAS cardioprotection by activating Gi-coupled S1P1 receptors (S1P1R) on MC. We report that activation of Gi-coupled S1P1R in cardiac MC confers IPC-like anti-RAS cardioprotection due to S1P1R-mediated inhibition of I/R-induced cardiac MC degranulation and renin release. This results from an initial translocation of protein kinase C subtype-ε and subsequent activation of aldehyde dehydrogenase type 2 (ALDH2), culminating in the elimination of the MC-degranulating effects of acetaldehyde and other toxic species produced during I/R. Inhibition of toxic aldehydes-induced MC-renin release prevents local RAS activation, reduces infarct size, and alleviates arrhythmias. Notably, these cardioprotective effects are lacking in hearts and MC from gene-targeted knock-in mice (ALDH2*2) in which ALDH2 enzymatic activity is maximally reduced. Thus, ALDH2 appears to play a pivotal role in this protective process. Our findings suggest that MC S1P1R may represent a new pharmacologic and therapeutic target for the direct alleviation of RAS-induced cardiac dysfunctions, including ischemic heart disease and congestive heart failure.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/physiology , Cardiotonic Agents/metabolism , Mast Cells/metabolism , Myocardial Reperfusion Injury/metabolism , Receptors, Lysosphingolipid/metabolism , Renin-Angiotensin System/physiology , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Gene Knock-In Techniques/methods , Guinea Pigs , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocardial Ischemia/metabolism , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Organ Culture Techniques , Swine
11.
Pharmacol Res ; 113(Pt A): 290-299, 2016 11.
Article in English | MEDLINE | ID: mdl-27616550

ABSTRACT

The endogenous gasotransmitter hydrogen sulphide (H2S) is an important regulator of the cardiovascular system, particularly of myocardial function. Moreover, H2S exhibits cardioprotective activity against ischemia/reperfusion (I/R) or hypoxic injury, and is considered an important mediator of "ischemic preconditioning", through activation of mitochondrial potassium channels, reduction of oxidative stress, activation of the endogenous "anti-oxidant machinery" and limitation of inflammatory responses. Accordingly, H2S-donors, i.e. pro-drugs able to generate exogenous H2S, are viewed as promising therapeutic agents for a number of cardiovascular diseases. The novel H2S-donor 4-carboxy phenyl-isothiocyanate (4CPI), whose vasorelaxing effects were recently reported, was tested here in different experimental models of myocardial I/R. In Langendorff-perfused rat hearts subjected to I/R, 4CPI significantly improved the post-ischemic recovery of myocardial functional parameters and limited tissue injury. These effects were antagonized by 5-hydroxydecanoic acid (a blocker of mitoKATP channels). Moreover, 4CPI inhibited the formation of reactive oxygen species. We found the whole battery of H2S-producing enzymes to be present in myocardial tissue: cystathionine γ-lyase (CSE), cystathionine ß-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (MPST). Notably, 4CPI down-regulated the post-ischemic expression of CSE. In Langendorff-perfused mouse hearts, 4CPI reduced the post-ischemic release of norepinephrine and the incidence of ventricular arrhythmias. In both rat and mouse hearts, 4CPI did not affect the degranulation of resident mast cells. In isolated rat cardiac mitochondria, 4CPI partially depolarized the mitochondrial membrane potential; this effect was antagonized by ATP (i.e., the physiological inhibitor of KATP channels). Moreover, 4CPI abrogated calcium uptake in the mitochondrial matrix. Finally, in an in vivo model of acute myocardial infarction in rats, 4CPI significantly decreased I/R-induced tissue injury. In conclusion, H2S-donors, and in particular isothiocyanate-based H2S-releasing drugs like 4CPI, can actually be considered a suitable pharmacological option in anti-ischemic therapy.


Subject(s)
Cardiotonic Agents/pharmacology , Hydrogen Sulfide/metabolism , Isothiocyanates/pharmacology , Myocardial Reperfusion Injury/drug therapy , Oxidative Stress/drug effects , Potassium Channels/metabolism , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cystathionine beta-Synthase/metabolism , Cystathionine beta-Synthase/pharmacology , Cystathionine gamma-Lyase/metabolism , Cysteine/analogs & derivatives , Cysteine/pharmacology , Decanoic Acids/pharmacology , Heart/drug effects , Hydroxy Acids/pharmacology , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
12.
Br J Pharmacol ; 173(22): 3222-3234, 2016 11.
Article in English | MEDLINE | ID: mdl-27548075

ABSTRACT

BACKGROUND AND PURPOSE: Hydrogen sulfide (H2 S) modulates many pathophysiological processes, including inflammation and allergic reactions, in which mast cells act as major effector cells. IgE receptor (FcεRI) cross linking leads to an increase in intracellular calcium ([Ca+2 ]i ), a critical step in mast cell degranulation. The aim of this study was to investigate the role of H2 S in [Ca+2 ]i -dependent mast cell activation. EXPERIMENTAL APPROACH: We investigated the effects of H2 S, either endogenously produced or released by the slow H2 S donor 4-carboxy-phenyl isothiocyanate (PhNCS-COOH), on antigenic- and non-antigenic degranulation of native murine mast cells, and human and rat (RBL-2H3) mast cell lines. We measured the release of specific mast cell degranulation markers (ß-hexosaminidase and renin), as well as changes in [Ca+2 ]i and phosphorylation of proteins downstream of FcεRI activation. KEY RESULTS: Endogenously produced H2 S inhibited antigen-induced degranulation in RBL-2H3. Similarly, H2 S released by PhNCS-COOH (10-300 µM) reduced, in a concentration-dependent manner, antigenic and non-antigenic degranulation and renin release in all mast cell types. Notably, PhNCS-COOH also prevented in a concentration-dependent mode the increase in [Ca+2 ]i elicited by Ca+2 ionophore, thapsigargin and FcεRI activation. Moreover, PhNCS-COOH attenuated the phosphorylation of Syk, cPLA-2 and PLCγ1 in antigen-stimulated RBL-2H3 cells. CONCLUSION AND IMPLICATIONS: Collectively, our results demonstrate that, by attenuating the phosphorylation of proteins downstream of FcεRI cross-linking on mast cells, H2 S diminishes [Ca+2 ]i availability and thus mast cell degranulation and renin release. These findings suggest that PhNCS-COOH could be a strategic therapeutic tool in mast cell-mediated allergic conditions.


Subject(s)
Benzoates/pharmacology , Calcium/metabolism , Cell Degranulation/drug effects , Hydrogen Sulfide/pharmacology , Isothiocyanates/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Renin/metabolism , Animals , Benzoates/chemistry , Hydrogen Sulfide/chemistry , Isothiocyanates/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Tumor Cells, Cultured
13.
FASEB J ; 29(1): 61-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25318477

ABSTRACT

Ischemia/reperfusion (I/R) elicits renin release from cardiac mast cells (MC), thus activating a local renin-angiotensin system (RAS), culminating in ventricular fibrillation. We hypothesized that in I/R, neurogenic ATP could degranulate juxtaposed MC and that ecto-nucleoside triphosphate diphosphohydrolase 1/CD39 (CD39) on MC membrane could modulate ATP-induced renin release. We report that pharmacological inhibition of CD39 in a cultured human mastocytoma cell line (HMC-1) and murine bone marrow-derived MC with ARL67156 (100 µM) increased ATP-induced renin release (≥2-fold), whereas purinergic P2X7 receptors (P2X7R) blockade with A740003 (3 µM) prevented it. Likewise, CD39 RNA silencing in HMC-1 increased ATP-induced renin release (≥2-fold), whereas CD39 overexpression prevented it. Acetaldehyde, an I/R product (300 µM), elicited an 80% increase in ATP release from HMC-1, in turn, causing an autocrine 20% increase in renin release. This effect was inhibited or potentiated when CD39 was overexpressed or silenced, respectively. Moreover, P2X7R silencing prevented ATP- and acetaldehyde-induced renin release. I/R-induced RAS activation in ex vivo murine hearts, characterized by renin and norepinephrine overflow and ventricular fibrillation, was potentiated (∼2-fold) by CD39 inhibition, an effect prevented by P2X7R blockade. Our data indicate that by regulating ATP availability at the MC surface, CD39 modulates local renin release and thus, RAS activation, ultimately exerting a cardioprotective effect.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Mast Cells/metabolism , Myocardial Reperfusion , Renin/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Antigens, CD/genetics , Apyrase/antagonists & inhibitors , Apyrase/genetics , Cardiotonic Agents/metabolism , Cell Degranulation , Cell Line , Humans , Male , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Myocardium/cytology , RNA, Small Interfering/genetics , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology
14.
Planta Med ; 80(8-9): 610-3, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24963613

ABSTRACT

Hydrogen sulfide is an endogenous pleiotropic gasotransmitter, which mediates important physiological effects in the human body. Accordingly, an impaired production of endogenous hydrogen sulfide contributes to the pathogenesis of important disorders. To date, exogenous compounds, acting as hydrogen sulfide-releasing agents, are viewed as promising pharmacotherapeutic agents. In a recent report, the hydrogen sulfide-releasing properties of some synthetic aryl isothiocyanate derivatives have been reported, indicating that the isothiocyanate function can be viewed as a suitable slow hydrogen sulfide-releasing moiety, endowed with the pharmacological potential typical of this gasotransmitter. Many isothiocyanate derivatives (deriving from a myrosinase-mediated transformation of glucosinolates) are well-known secondary metabolites of plants belonging to the family Brassicaceae, a large botanical family comprising many edible species. The phytotherapeutic and nutraceutic usefulness of Brassicaceae in the prevention of important human diseases, such as cancer, neurodegenerative processes and cardiovascular diseases has been widely discussed in the scientific literature. Although these effects have been largely attributed to isothiocyanates, the exact mechanism of action is still unknown. In this experimental work, we aimed to investigate the possible hydrogen sulfide-releasing capacity of some important natural isothiocyanates, studying it in vitro by amperometric detection. Some of the tested natural isothiocyanates exhibited significant hydrogen sulfide release, leading us to hypothesize that hydrogen sulfide may be, at least in part, a relevant player accounting for several biological effects of Brassicaceae.


Subject(s)
Brassicaceae/chemistry , Gasotransmitters/pharmacokinetics , Hydrogen Sulfide/pharmacokinetics , Isothiocyanates/pharmacology , Gasotransmitters/chemistry , Glucosinolates/chemistry , Glucosinolates/pharmacology , Humans , Hydrogen Sulfide/chemistry , Isothiocyanates/chemistry
15.
J Pharmacol Exp Ther ; 349(3): 508-17, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696042

ABSTRACT

Renin released by ischemia/reperfusion (I/R) from cardiac mast cells (MCs) activates a local renin-angiotensin system (RAS) causing arrhythmic dysfunction. Ischemic preconditioning (IPC) inhibits MC renin release and consequent activation of this local RAS. We postulated that MC histamine H4-receptors (H4Rs), being Gαi/o-coupled, might activate a protein kinase C isotype-ε (PKCε)-aldehyde dehydrogenase type-2 (ALDH2) cascade, ultimately eliminating MC-degranulating and renin-releasing effects of aldehydes formed in I/R and associated arrhythmias. We tested this hypothesis in ex vivo hearts, human mastocytoma cells, and bone marrow-derived MCs from wild-type and H4R knockout mice. We found that activation of MC H4Rs mimics the cardioprotective anti-RAS effects of IPC and that protection depends on the sequential activation of PKCε and ALDH2 in MCs, reducing aldehyde-induced MC degranulation and renin release and alleviating reperfusion arrhythmias. These cardioprotective effects are mimicked by selective H4R agonists and disappear when H4Rs are pharmacologically blocked or genetically deleted. Our results uncover a novel cardioprotective pathway in I/R, whereby activation of H4Rs on the MC membrane, possibly by MC-derived histamine, leads sequentially to PKCε and ALDH2 activation, reduction of toxic aldehyde-induced MC renin release, prevention of RAS activation, reduction of norepinephrine release, and ultimately to alleviation of reperfusion arrhythmias. This newly discovered protective pathway suggests that MC H4Rs may represent a new pharmacologic and therapeutic target for the direct alleviation of RAS-induced cardiac dysfunctions, including ischemic heart disease and congestive heart failure.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Ischemic Preconditioning , Mast Cells/metabolism , Myocardial Reperfusion Injury/metabolism , Protein Kinase C-epsilon/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Renin/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Animals , Cell Differentiation , Cell Line , Enzyme Activation , Guinea Pigs , Humans , In Vitro Techniques , Mast Cells/enzymology , Mice , Mice, Knockout , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/genetics , Receptors, Histamine H4 , Renin-Angiotensin System/physiology
16.
Vascul Pharmacol ; 60(1): 32-41, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24287004

ABSTRACT

Hydrogen sulfide (H2S) is an endogenous gasotransmitter, which mediates important physiological effects in the cardiovascular system. Accordingly, an impaired production of endogenous H2S contributes to the pathogenesis of important cardiovascular disorders, such as hypertension. Therefore, exogenous compounds, acting as H2S-releasing agents, are viewed as promising pharmacotherapeutic agents for cardiovascular diseases. Thus, this paper aimed at evaluating the H2S-releasing properties of some aryl isothiocyanate derivatives and their vascular effects. The release of H2S was determined by amperometry, spectrophotometry and gas/mass chromatography. Moreover, the vascular activity of selected isothiocyanates were tested in rat conductance (aorta) and coronary arteries. Since H2S has been recently reported to act as an activator of vascular Kv7 potassium channels, the possible membrane hyperpolarizing effects of isothiocyanates were tested on human vascular smooth muscle (VSM) cells by spectrofluorescent dyes. Among the tested compounds, phenyl isothiocyanate (PhNCS) and 4-carboxyphenyl isothiocyanate (PhNCS-COOH) exhibited slow-H2S-release, triggered by organic thiols such as L-cysteine. These compounds were endowed with vasorelaxing effects on conductance and coronary arteries. Moreover, these two isothiocyanates caused membrane hyperpolarization of VSM cells. The vascular effects of isothiocyanates were strongly abolished by the selective Kv7-blocker XE991. In conclusion, the isothiocyanate function can be viewed as a suitable slow H2S-releasing moiety, endowed with vasorelaxing and hypotensive effects, typical of this gasotransmitter. Thus, such a chemical moiety can be employed for the development of novel chemical tools for basic studies and promising cardiovascular drugs.


Subject(s)
Hydrogen Sulfide/pharmacology , Isothiocyanates/pharmacology , Muscle, Smooth, Vascular/drug effects , Animals , Aorta/drug effects , Aorta/physiology , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Muscle, Smooth, Vascular/physiology , Rats , Rats, Wistar , Vasoconstriction/drug effects , Vasodilation/drug effects
17.
ACS Med Chem Lett ; 4(10): 904-8, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24900583

ABSTRACT

A small library of arylthioamides 1-12 was easily synthesized, and their H2S-releasing properties were evaluated both in the absence or in the presence of an organic thiol such as l-cysteine. A number of arylthioamides (1-3 and 7) showed a slow and l-cysteine-dependent H2S-releasing mechanism, similar to that exhibited by the reference slow H2S-releasing agents, such as diallyl disulfide (DADS) and the phosphinodithioate derivative GYY 4137. Compound 1 strongly abolished the noradrenaline-induced vasoconstriction in isolated rat aortic rings and hyperpolarized the membranes of human vascular smooth muscle cells in a concentration-dependent fashion. Finally, a significant reduction of the systolic blood pressure of anesthetized normotensive rats was observed after its oral administration. Altogether these results highlighted the potential of arylthioamides 1-3 and 7 as H2S-donors for basic studies, and for the rational design/development of promising pharmacotherapeutic agents to treat cardiovascular diseases.

SELECTION OF CITATIONS
SEARCH DETAIL
...