Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38542360

ABSTRACT

Different gut microbiota-derived metabolites influence cardiovascular function, and, among all, the role of indole-3-propionic acid (IPA), from tryptophan metabolism, shows controversial effects. The aim of this study was to evaluate its role in endothelial dysfunction. IPA effects were studied on bovine aortic endothelial cells (BAE-1). First, IPA cytotoxicity was evaluated by an MTS assay. Then, the levels of intracellular reactive oxygen species (ROS) were evaluated by a microplate reader or fluorescence microscopy with the CellROX® Green probe, and nitric oxide (NO) production was studied by fluorescence microscopy with the DAR4M-AM probe after acute or chronic treatment. Finally, immunoblotting analysis for endothelial nitric oxide synthase (eNOS) phosphorylation (p-eNOS) was performed. In BAE-1, IPA was not cytotoxic, except for the highest concentration (5 mM) after 48 h of treatment, and it showed neither oxidant nor antioxidant activity. However, the physiological concentration of IPA (1 µM) significantly reduced NO released by adenosine triphosphate (ATP)-stimulated BAE-1. These last data were confirmed by Western blot analysis, where IPA induced a significant reduction in p-eNOS in purinergic-stimulated BAE-1. Given these data, we can speculate that IPA negatively affects the physiological control of vascular tone by impairing the endothelial NO release induced by purinergic stimulation. These results represent a starting point for understanding the mechanisms underlying the relationship between gut microbiota metabolites and cardiometabolic health.


Subject(s)
Gastrointestinal Microbiome , Propionates , Vascular Diseases , Animals , Cattle , Endothelial Cells/metabolism , Nitric Oxide/metabolism , Tryptophan/metabolism , Vascular Diseases/metabolism , Nitric Oxide Synthase Type III/metabolism , Indoles/pharmacology , Indoles/metabolism
2.
Food Funct ; 14(9): 4163-4172, 2023 May 11.
Article in English | MEDLINE | ID: mdl-37062967

ABSTRACT

A healthy vascular endothelium plays an essential role in modulating vascular tone by producing and releasing vasoactive factors such as nitric oxide (NO). Endothelial dysfunction (ED), the loss of the endothelium physiological functions, results in the inability to properly regulate vascular tone, leading to hypertension and other cardiovascular risk factors. Alongside NO, the gasotransmitter hydrogen sulfide (H2S) has emerged as a key molecule with vasodilatory and antioxidant activities. Since a reduction in H2S bioavailability is related to ED pathogenesis, natural H2S donors are very attractive. In particular, we focused on the sulfur-containing amino acid S-allyl cysteine (SAC), a bioactive metabolite, of which black garlic is particularly rich, with antioxidant activity and, among others, anti-diabetic and anti-hypertensive properties. In this study, we analyzed the protective effect of SAC against ED by evaluating reactive oxygen species level, H2S release, eNOS phosphorylation, and NO production (by fluorescence imaging and western blot analysis) in Bovine Aortic Endothelial cells (BAE-1). Furthermore, we chemically characterized a Black Garlic Extract (BGE) for its content in SAC and other sulfur-containing amino acids. BGE was used to carry out an analysis on H2S release on BAE-1 cells. Our results show that both SAC and BGE significantly increase H2S release. Moreover, SAC reduces ROS production and enhances eNOS phosphorylation and the consequent NO release in our cellular model. In this scenario, a natural extract enriched in SAC could represent a novel therapeutic approach to prevent the onset of ED-related diseases.


Subject(s)
Garlic , Hydrogen Sulfide , Animals , Cattle , Antioxidants/pharmacology , Antioxidants/metabolism , Sulfur Compounds/pharmacology , Garlic/chemistry , Endothelial Cells/metabolism , Hydrogen Sulfide/metabolism , Cysteine/pharmacology , Endothelium, Vascular/metabolism , Sulfur
3.
Int J Mol Sci ; 24(6)2023 Mar 18.
Article in English | MEDLINE | ID: mdl-36982880

ABSTRACT

Endothelial function is essential in the maintenance of systemic homeostasis, whose modulation strictly depends on the proper activity of tissue-specific angiocrine factors on the physiopathological mechanisms acting at both single and multi-organ levels. Several angiocrine factors take part in the vascular function itself by modulating vascular tone, inflammatory response, and thrombotic state. Recent evidence has outlined a strong relationship between endothelial factors and gut microbiota-derived molecules. In particular, the direct involvement of trimethylamine N-oxide (TMAO) in the development of endothelial dysfunction and its derived pathological outcomes, such as atherosclerosis, has come to light. Indeed, the role of TMAO in the modulation of factors strictly related to the development of endothelial dysfunction, such as nitric oxide, adhesion molecules (ICAM-1, VCAM-1, and selectins), and IL-6, has been widely accepted. The aim of this review is to present the latest studies that describe a direct role of TMAO in the modulation of angiocrine factors primarily involved in the development of vascular pathologies.


Subject(s)
Gastrointestinal Microbiome , Vascular Diseases , Humans , Gastrointestinal Microbiome/physiology , Methylamines/metabolism
4.
Int J Mol Sci ; 23(7)2022 Apr 02.
Article in English | MEDLINE | ID: mdl-35409341

ABSTRACT

Trimethylamine N-oxide (TMAO) is a diet derived compound directly introduced through foodstuff, or endogenously synthesized from its precursors, primarily choline, L-carnitine, and ergothioneine. New evidence outlines high TMAO plasma concentrations in patients with overt cardiovascular disease, but its direct role in pathological development is still controversial. The purpose of the study was to evaluate the role of TMAO in affecting key intracellular factors involved in endothelial dysfunction development, such as reactive oxygen species, mitochondrial health, calcium balance, and nitric oxide release using bovine aortic endothelial cells (BAE-1). Cell viability and oxidative stress indicators were monitored after acute and prolonged TMAO treatment. The role of TMAO in interfering with the physiological purinergic vasodilatory mechanism after ATP stimulation was defined through measurements of the rise of intracellular calcium, nitric oxide release, and eNOS phosphorylation at Ser1179 (eNOSSer1179). TMAO was not cytotoxic for BAE-1 and it did not induce the rise of reactive oxygen species and impairment of mitochondrial membrane potential, either in the basal condition or in the presence of a stressor. In contrast, TMAO modified the purinergic response affecting intracellular ATP-induced calcium increase, nitric oxide release, and eNOSSer1179. Results obtained suggest a possible implication of TMAO in impairing the endothelial-dependent vasodilatory mechanism.


Subject(s)
Calcium , Nitric Oxide , Adenosine Triphosphate , Animals , Calcium, Dietary , Cattle , Endothelial Cells/metabolism , Humans , Methylamines , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism
5.
Vascul Pharmacol ; 141: 106921, 2021 12.
Article in English | MEDLINE | ID: mdl-34592428

ABSTRACT

Ischemic Heart Disease (IHD) is a clinical condition characterized by insufficient blood flow to the cardiac tissue, and the consequent inappropriate oxygen and nutrients supply and metabolic waste removal in the heart. In the last decade a broad scientific literature has underlined the distinct mechanism of onset and the peculiar progress of IHD between female and male patients, highlighting the estrogenic hormonal setting as a key factor of these sex-dependent divergences. In particular, estrogen-activated cardioprotective pathways exert a pivotal role for the microvascular health, and their impairment, both physiologically and pathologically driven, predispose to vascular dysfunctions. Aim of this review is to summarize the current knowledge on the estrogen receptors localization and function in the cardiovascular system, particularly focusing on sex-dependent differences in microvascular vs macrovascular dysfunction and on the experimental models that allowed the researchers to reach the current findings and sketching the leading estrogen-mediated cardioprotective mechanisms.


Subject(s)
Myocardial Ischemia , Estrogens , Estrone , Female , Heart , Humans , Male , Myocardial Ischemia/drug therapy , Myocardial Ischemia/prevention & control , Receptors, Estrogen
6.
Front Physiol ; 12: 667961, 2021.
Article in English | MEDLINE | ID: mdl-34054579

ABSTRACT

Ischemic heart disease (IHD) is a multifactorial pathological condition strictly related to genetic, dietary, and lifestyle factors. Its morbidity and mortality rate represent one of the most important pathological issues that today involve younger people in a stronger way than in the past. IHD clinical outcomes are difficult to treat and have a high economic impact on health care. So prevention of this pathological condition through cardioprotective maneuvers represents the first line of intervention, as already underlined by several animal and human studies. Even if the time of intervention is important to prevent severe outcomes, many studies highlight that sex-dependent responses are crucial for the result of cardioprotective procedures. In this scenario sexual hormones have revealed an important role in cardioprotective approach, as women seem to be more protected toward cardiac insults when compared to male counterparts. The aim of this mini review is to show the molecular pathways involved in cardioprotective protocols and to elucidate how sexual hormones can contribute in ameliorating or worsening the physiological responses to IHD.

7.
Nutrients ; 13(3)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809114

ABSTRACT

Skeletal muscle plays a pivotal role in whole-body glucose metabolism, accounting for the highest percentage of glucose uptake and utilization in healthy subjects. Impairment of these key functions occurs in several conditions including sedentary lifestyle and aging, driving toward hyperglycemia and metabolic chronic diseases. Therefore, strategies pointed to improve metabolic health by targeting skeletal muscle biochemical pathways are extremely attractive. Among them, we focused on the natural sesquiterpene and cannabinoid type 2 (CB2) receptor agonist Trans-ß-caryophyllene (BCP) by analyzing its role in enhancing glucose metabolism in skeletal muscle cells. Experiments were performed on C2C12 myotubes. CB2 receptor membrane localization in myotubes was assessed by immunofluorescence. Within glucose metabolism, we evaluated glucose uptake (by the fluorescent glucose analog 2-NBDG), key enzymes of both glycolytic and oxidative pathways (by spectrophotometric assays and metabolic radiolabeling) and ATP production (by chemiluminescence-based assays). In all experiments, CB2 receptor involvement was tested with the CB2 antagonists AM630 and SR144528. Our results show that in myotubes, BCP significantly enhances glucose uptake, glycolytic and oxidative pathways, and ATP synthesis through a CB2-dependent mechanism. Giving these outcomes, CB2 receptor stimulation by BCP could represent an appealing tool to improve skeletal muscle glucose metabolism, both in physiological and pathological conditions.


Subject(s)
Adenosine Triphosphate/biosynthesis , Glucose/metabolism , Muscle, Skeletal/drug effects , Plant Extracts/pharmacology , Polycyclic Sesquiterpenes/pharmacology , Receptor, Cannabinoid, CB2/agonists , Animals , Cell Line , Electron Transport/drug effects , Fluorescent Antibody Technique , Glycolysis/drug effects , Mice , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Piper nigrum , Receptor, Cannabinoid, CB2/drug effects
8.
Nutrients ; 12(11)2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33114564

ABSTRACT

(E)-ß-caryophyllene (BCP) is a bicyclic sesquiterpene widely distributed in the plant kingdom, where it contributes a unique aroma to essential oils and has a pivotal role in the survival and evolution of higher plants. Recent studies provided evidence for protective roles of BCP in animal cells, highlighting its possible use as a novel therapeutic tool. Experimental results show the ability of BCP to reduce pro-inflammatory mediators such as tumor necrosis factor-alfa (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), thus ameliorating chronic pathologies characterized by inflammation and oxidative stress, in particular metabolic and neurological diseases. Through the binding to CB2 cannabinoid receptors and the interaction with members of the family of peroxisome proliferator-activated receptors (PPARs), BCP shows beneficial effects on obesity, non-alcoholic fatty liver disease/nonalcoholic steatohepatitis (NAFLD/NASH) liver diseases, diabetes, cardiovascular diseases, pain and other nervous system disorders. This review describes the current knowledge on the biosynthesis and natural sources of BCP, and reviews its role and mechanisms of action in different inflammation-related metabolic and neurologic disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chronic Disease/drug therapy , Inflammation/drug therapy , Plant Extracts/pharmacology , Polycyclic Sesquiterpenes/pharmacology , Animals , Humans , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , PPAR gamma/metabolism , Receptor, Cannabinoid, CB2/metabolism
9.
Antioxidants (Basel) ; 9(8)2020 Aug 02.
Article in English | MEDLINE | ID: mdl-32748847

ABSTRACT

Squalene (SQ) is a natural triterpene widely distributed in nature. It is a metabolic intermediate of the sterol biosynthetic pathway and represents a possible target in different metabolic and oxidative stress-related disorders. Growing interest has been focused on SQ's antioxidant properties, derived from its chemical structure. Strong evidence provided by ex vivo models underline its scavenging activity towards free radicals, whereas only a few studies have highlighted its effect in cellular models of oxidative stress. Given the role of unbalanced free radicals in both the onset and progression of several cardiovascular diseases, an in depth evaluation of SQ's contribution to antioxidant defense mechanisms could represent a strategic approach in dealing with these pathological conditions. At present experimental results overall show a double-edged sword role of squalene in cardiovascular diseases and its function has to be better elucidated in order to establish intervention lines focused on its features. This review aims to summarize current knowledge about endogenous and exogenous sources of SQ and to point out the controversial role of SQ in cardiovascular physiology.

10.
Nutrients ; 11(11)2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31731718

ABSTRACT

Trans-ß-caryophyllene (BCP) is a natural sesquiterpene hydrocarbon with several important pharmacological activities, including antioxidant, anti-inflammatory, anticancer, and cardioprotective functions. These properties are mainly due to its selective interaction with the peripherally expressed cannabinoid receptor 2. In addition, BCP activates peroxisome proliferated activator receptors α and γ and inhibits the Toll-like receptor signaling pathway. Given the growing scientific interest in BCP, the aim of our study was to investigate the metabolic effects of a black pepper extract (PipeNig®-FL), containing a high standardized content of BCP. In particular our interest was focused on its potential activity on lipid accumulation and glucose uptake. The extract PipeNig®-FL was chemically characterized by gas chromatography-mass spectrometry (GC-MS) and gas chromatography with flame-ionization detection (GC-FID), confirming a high content (814 mg/g) of BCP. Experiments were performed on 3T3-L1 preadipocytes and on C2C12 myotubes. Lipid content following 3T3-L1 adipogenic differentiation was quantified with AdipoRed fluorescence staining. Glucose uptake and GLUT4 membrane translocation were studied in C2C12 myotubes with the fluorescent glucose analog 2-NBDG and by immunofluorescence analysis. Here we show that PipeNig®-FL reduces 3T3-L1 adipocyte differentiation and lipid accumulation. Moreover, acute exposure of C2C12 myotubes to PipeNig®-FL improves glucose uptake activity and GLUT4 migration. Taken together, these results reveal interesting and novel properties of BCP, suggesting potential applications in the prevention of lipid accumulation and in the improvement of glucose uptake.


Subject(s)
Lipid Metabolism/drug effects , Piper nigrum/chemistry , Plant Extracts/pharmacology , Polycyclic Sesquiterpenes/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/drug effects , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gas Chromatography-Mass Spectrometry , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Mice , Muscle Fibers, Skeletal/drug effects , Plant Extracts/chemistry
11.
Int J Mol Sci ; 20(12)2019 Jun 21.
Article in English | MEDLINE | ID: mdl-31234461

ABSTRACT

Trimethylamine N-oxide (TMAO) is an organic compound derived from dietary choline and L-carnitine. It behaves as an osmolyte, a protein stabilizer, and an electron acceptor, showing different biological functions in different animals. Recent works point out that, in humans, high circulating levels of TMAO are related to the progression of atherosclerosis and other cardiovascular diseases. However, studies on a direct role of TMAO in cardiomyocyte parameters are still limited. The purpose of this work is to study the effects of TMAO on isolated adult rat cardiomyocytes. TMAO in both 100 µM and 10 mM concentrations, from 1 to 24 h of treatment, does not affect cell viability, sarcomere length, intracellular ROS, and mitochondrial membrane potential. Furthermore, the simultaneous treatment with TMAO and known cardiac insults, such as H2O2 or doxorubicin, does not affect the treatment's effect. In conclusion, TMAO cannot be considered a direct cause or an exacerbating risk factor of cardiac damage at the cellular level in acute conditions.


Subject(s)
Membrane Potential, Mitochondrial , Methylamines/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Survival , Cells, Cultured , Female , Myocytes, Cardiac/cytology , Rats
12.
Biomed Res Int ; 2018: 2086109, 2018.
Article in English | MEDLINE | ID: mdl-30370303

ABSTRACT

Catestatin is a cationic and hydrophobic peptide derived from the enzymatic cleavage of the prohormone Chromogranin A. Initially identified as a potent endogenous nicotinic-cholinergic antagonist, Catestatin has recently been shown to act as a novel regulator of cardiac function and blood pressure and as a cardioprotective agent in both pre- and postconditioning through AKT-dependent mechanisms. The aim of this study is to investigate the potential role of Catestatin also on cardiac metabolism modulation, particularly on cardiomyocytes glucose uptake. Experiments were performed on isolated adult rat cardiomyocytes. Glucose uptake was assessed by fluorescent glucose incubation and confocal microscope analysis. Glut4 plasma membrane translocation was studied by immunofluorescence experiments and evaluation of the ratio peripheral vs internal Glut4 staining. Furthermore, we performed immunoblot experiments to investigate the involvement of the intracellular pathway AKT/AS160 in the Catestatin dependent Glut4 trafficking. Our results show that 10 nM Catestatin induces a significant increase in the fluorescent glucose uptake, comparable to that exerted by 100 nM Insulin. Moreover, Catestatin stimulates Glut4 translocation to plasma membrane and both AKT and AS160 phosphorylation. All these effects were inhibited by Wortmannin. On the whole, we show for the first time that Catestatin is able to modulate cardiac glucose metabolism, by inducing an increase in glucose uptake through Glut4 translocation to the plasma membrane and that this mechanism is mediated by the AKT/AS160 intracellular pathway.


Subject(s)
Chromogranin A/pharmacology , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Myocytes, Cardiac/drug effects , Peptide Fragments/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , GTPase-Activating Proteins/metabolism , Insulin/metabolism , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats
13.
Proc Natl Acad Sci U S A ; 114(45): 12033-12038, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078377

ABSTRACT

It has been shown that growth hormone-releasing hormone (GHRH) reduces cardiomyocyte (CM) apoptosis, prevents ischemia/reperfusion injury, and improves cardiac function in ischemic rat hearts. However, it is still not known whether GHRH would be beneficial for life-threatening pathological conditions, like cardiac hypertrophy and heart failure (HF). Thus, we tested the myocardial therapeutic potential of GHRH stimulation in vitro and in vivo, using GHRH or its agonistic analog MR-409. We show that in vitro, GHRH(1-44)NH2 attenuates phenylephrine-induced hypertrophy in H9c2 cardiac cells, adult rat ventricular myocytes, and human induced pluripotent stem cell-derived CMs, decreasing expression of hypertrophic genes and regulating hypertrophic pathways. Underlying mechanisms included blockade of Gq signaling and its downstream components phospholipase Cß, protein kinase Cε, calcineurin, and phospholamban. The receptor-dependent effects of GHRH also involved activation of Gαs and cAMP/PKA, and inhibition of increase in exchange protein directly activated by cAMP1 (Epac1). In vivo, MR-409 mitigated cardiac hypertrophy in mice subjected to transverse aortic constriction and improved cardiac function. Moreover, CMs isolated from transverse aortic constriction mice treated with MR-409 showed improved contractility and reversal of sarcolemmal structure. Overall, these results identify GHRH as an antihypertrophic regulator, underlying its therapeutic potential for HF, and suggest possible beneficial use of its analogs for treatment of pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Growth Hormone-Releasing Hormone/metabolism , Heart Failure/metabolism , Heart/physiology , Animals , Apoptosis/drug effects , Calcineurin/metabolism , Cardiomegaly/chemically induced , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phenylephrine/pharmacology , Phospholipase C beta/metabolism , Protein Kinase C/metabolism , Rats , Signal Transduction/drug effects
14.
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
15.
Eur J Med Chem ; 113: 187-97, 2016 May 04.
Article in English | MEDLINE | ID: mdl-26942626

ABSTRACT

We report the synthesis and characterization of a series of symmetrical indolenine-based squaraine dyes along with the evaluation of their singlet oxygen generation efficiency. The photodynamic activity of these new photosensitizers has been evaluated on a human tumor fibrosarcoma (HT-1080) cell line. The cytotoxicity increased over time and is induced by the photoactivation of bromo (Br-C4) and iodio (I-C4) long carbon chain squaraine dyes and the consequent increase in reactive oxygen species (ROS) production (p < 0.001), which leads to necrosis 6 h after treatment. Induction of cytochrome c release, DNA damage and up-regulation of GPX1, NQO1 and SOD2 mRNA gene expression after PDT were investigated.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclobutanes/pharmacology , Halogens/chemistry , Phenols/pharmacology , Photosensitizing Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Cyclobutanes/chemical synthesis , Cyclobutanes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Phenols/chemical synthesis , Phenols/chemistry , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
16.
Curr Drug Targets ; 16(8): 843-67, 2015.
Article in English | MEDLINE | ID: mdl-25751010

ABSTRACT

Cardiovascular diseases (CVD) are the leading cause of death, chronic illness and disability in Western countries. The most common cause of CVD derives from the harmful effects of acute myocardial ischemia and subsequent reperfusion injury. Cardioprotection against acute ischemia/ reperfusion injury is made possible by the "conditioning protocols." Conditioning is obtained by applying a few periods of brief ischemia and reperfusion in the event of prolonged (index) ischemia that may cause myocardial infarction. Whilst the conditioning stimulus is applied before the index ischemia in ischemic pre-conditioning, it is applied after the event in post-conditioning. Pre and post- conditioning stimuli can be applied in a different/remote organ (remote pre- and post-conditioning); in this case conditioning stimulus can also be applied during the index event, in the so called remote per-conditioning. All these endogenous cardioprotective strategies recruit endogenous cytoprotective agents and factors that elicit specific cardioprotective pathways. Here, we discuss many of these cardioprotective factors compared to literature and highlight their main characteristics and mechanisms of action. Enphasis is given to endogenous cardioprotective agents acting or not on surface receptors, including chromogranin A derivatives, ghrelin-associated peptides, growth factors and cytokines, and to microvesicles and exosomes. Moreover the cardioprotective effects of gasotransmitters nitric oxide, hydrogen sulphide and carbon monoxide are reviewed. The possible clinical translation of these knowledge for future successful therapies is briefly and critically discussed.


Subject(s)
Cardiotonic Agents/metabolism , Ischemic Postconditioning/methods , Ischemic Preconditioning, Myocardial/methods , Myocardial Ischemia/prevention & control , Myocardial Reperfusion Injury/prevention & control , Animals , Carbon Monoxide/metabolism , Gasotransmitters/metabolism , Humans , Hydrogen Sulfide/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/therapy , Nitric Oxide/metabolism
17.
PLoS One ; 10(3): e0119790, 2015.
Article in English | MEDLINE | ID: mdl-25774921

ABSTRACT

Catestatin (Cst) is a 21-amino acid peptide deriving from Chromogranin A. Cst exerts an overall protective effect against an excessive sympathetic stimulation of cardiovascular system, being able to antagonize catecholamine secretion and to reduce their positive inotropic effect, by stimulating the release of nitric oxide (NO) from endothelial cells. Moreover, Cst reduces ischemia/reperfusion (I/R) injury, improving post-ischemic cardiac function and cardiomyocyte survival. To define the cardioprotective signaling pathways activated by Cst (5 nM) we used isolated adult rat cardiomyocytes undergoing simulated I/R. We evaluated cell viability rate with propidium iodide labeling and mitochondrial membrane potential (MMP) with the fluorescent probe JC-1. The involvement of Akt, GSK3ß, eNOS and phospholamban (PLN) cascade was studied by immunofluorescence. The role of PI3K-Akt/NO/cGMP pathway was also investigated by using the pharmacological blockers wortmannin (Wm), L-NMMA and ODQ. Our experiments revealed that Cst increased cell viability rate by 65% and reduced cell contracture in I/R cardiomyocytes. Wm, L-NMMA and ODQ limited the protective effect of Cst. The protective outcome of Cst was related to its ability to maintain MMP and to increase AktSer473, GSK3ßSer9, PLNThr17 and eNOSSer1179 phosphorylation, while treatment with Wm abolished these effects. Thus, the present results show that Cst is able to exert a direct action on cardiomyocytes and give new insights into the molecular mechanisms involved in its protective effect, highlighting the PI3K/NO/cGMP pathway as the trigger and the MMP preservation as the end point of its action.


Subject(s)
Chromogranin A/pharmacology , Membrane Potential, Mitochondrial/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Chromogranin A/therapeutic use , Glycogen Synthase Kinase 3/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Peptide Fragments/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats
18.
Biomed Res Int ; 2014: 783623, 2014.
Article in English | MEDLINE | ID: mdl-25136621

ABSTRACT

The chromogranin-A peptide catestatin modulates a wide range of processes, such as cardiovascular functions, innate immunity, inflammation, and metabolism. We recently found that the cardiac antiadrenergic action of catestatin requires a PI3K-dependent NO release from endothelial cells, although the receptor involved is yet to be identified. In the present work, based on the cationic properties of catestatin, we tested the hypothesis of its interaction with membrane heparan sulphate proteoglycans, resulting in the activation of a caveolae-dependent endocytosis. Experiments were performed on bovine aortic endothelial cells. Endocytotic vesicles trafficking was quantified by confocal microscopy using a water-soluble membrane dye; catestatin colocalization with heparan sulphate proteoglycans and caveolin 1 internalization were studied by fluorimetric measurements in live cells. Modulation of the catestatin-dependent eNOS activation was assessed by immunofluorescence and immunoblot analysis. Our results demonstrate that catestatin (5 nM) colocalizes with heparan sulphate proteoglycans and induces a remarkable increase in the caveolae-dependent endocytosis and caveolin 1 internalization, which were significantly reduced by both heparinase and wortmannin. Moreover, catestatin was unable to induce Ser(1179) eNOS phosphorylation after pretreatments with heparinase and methyl-ß-cyclodextrin. Taken together, these results highlight the obligatory role for proteoglycans and caveolae internalization in the catestatin-dependent eNOS activation in endothelial cells.


Subject(s)
Chromogranin A/administration & dosage , Endocytosis/drug effects , Heparan Sulfate Proteoglycans/metabolism , Nitric Oxide/metabolism , Peptide Fragments/administration & dosage , Animals , Aorta/drug effects , Aorta/metabolism , Cattle , Caveolae/drug effects , Caveolae/ultrastructure , Caveolin 1/metabolism , Chromogranin A/metabolism , Chromogranin A/ultrastructure , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure
19.
Diabetes ; 63(7): 2380-93, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24622796

ABSTRACT

RFamide peptides 43RFa and 26RFa have been shown to promote food intake and to exert different peripheral actions through G-protein-coupled receptor 103 (GPR103) binding. Moreover, 26RFa was found to inhibit pancreatic insulin secretion, whereas the role of 43RFa on ß-cell function is unknown, as well as the effects of both peptides on ß-cell survival. Herein, we investigated the effects of 43RFa and 26RFa on survival and apoptosis of pancreatic ß-cells and human pancreatic islets. In addition, we explored the role of these peptides on insulin secretion and the underlying signaling mechanisms. Our results show that in INS-1E ß-cells and human pancreatic islets both 43RFa and 26RFa prevented cell death and apoptosis induced by serum starvation, cytokine synergism, and glucolipotoxicity, through phosphatidylinositol 3-kinase/Akt- and extracellular signal-related kinase 1/2-mediated signaling. Moreover, 43RFa promoted, whereas 26RFa inhibited, glucose- and exendin-4-induced insulin secretion, through Gαs and Gαi/o proteins, respectively. Inhibition of GPR103 expression by small interfering RNA blocked 43RFa insulinotropic effect, but not the insulinostatic action of 26RFa. Finally, 43RFa, but not 26RFa, induced cAMP increase and glucose uptake. In conclusion, because of their survival effects along with the effects on insulin secretion, these findings suggest potential for 43RFa and 26RFa as therapeutic targets in the treatment of diabetes.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin/metabolism , Islets of Langerhans/drug effects , Neuropeptides/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Insulin Secretion , Insulin-Secreting Cells/physiology , Intercellular Signaling Peptides and Proteins , Islets of Langerhans/physiology , Peptides/genetics , Peptides/metabolism , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
20.
J Cell Biochem ; 113(3): 800-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22020979

ABSTRACT

In recent years cardiac tissue engineering has emerged as a promising field aimed at developing suitable techniques to repair the infarcted myocardium with a combination of cells, biomaterials, and regulative factors. In particular it could stand for an alternative strategy to simple in situ cellular implantation. In the present study our purpose was to analyze the interaction between a hyaluronan-based mesh (HYALONECT®) and neonatal murine ventricular myocytes (NMVMs). Specifically, we investigated morphological and functional characteristics of cardiomyocytes cultured on HYALONECT® in view of its employment in heart repair. Both living and fixed cells analysis was performed on in toto scaffolds with confocal microscopy. NMVMs adhesion on HYALONECT® was studied by tracking sarcomeric α-actinin immunofluorescence staining. The structural features of NMVMs adherent onto HYALONECT® were investigated at 24, 48, 72 h, and 7 days of culture by immunofluorescence for sarcomeric α-actinin and connexin-43. We observed a progressive morphological organization of the cells inside the biopolymer, with both clear sarcomeric arrangement along the scaffold fibers and gap junctions development between adjacent cells. Finally, in vivo intracellular calcium measurements performed using calcium fluorimetric confocal imaging revealed the presence of spontaneous calcium transients and contractile activity of NMVMs adherent onto HYALONECT® up to 48 h from seeding, indicating a progressive differentiation of the cells toward the adult phenotype. In conclusion, our results demonstrate that HYALONECT® allowed NMVMs to adhere to the fibers and to develop functional properties, displaying suitable features as a scaffold to perform heart tissue engineering.


Subject(s)
Hyaluronic Acid , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Tissue Scaffolds , Animals , Animals, Newborn , Cell Adhesion , Cell Separation , Cells, Cultured , Heart Ventricles/cytology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...