Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 23(17)2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36077595

ABSTRACT

The present study investigates infarct-reducing effects of blocking ischemia-induced opening of connexin43 hemichannels using peptides Gap19, Gap26 or Gap27. Cardioprotection by ischemic preconditioning (IPC) and Gap peptides was compared, and combined treatment was tested in isolated, perfused male rat hearts using function and infarct size after global ischemia, high-resolution respirometry of isolated mitochondrial and peptide binding kinetics as endpoints. The Gap peptides reduced infarct size significantly when given prior to ischemia plus at reperfusion (Gap19 76.2 ± 2.7, Gap26 72.9 ± 5.8 and Gap27 71.9 ± 5.8% of untreated control infarcts, mean ± SEM). Cardioprotection was lost when Gap26, but not Gap27 or Gap19, was combined with triggering IPC (IPC 73.4 ± 5.5, Gap19-IPC 60.9 ± 5.1, Gap26-IPC 109.6 ± 7.8, Gap27-IPC 56.3 ± 8.0% of untreated control infarct). Binding stability of peptide Gap26 to its specific extracellular loop sequence (EL2) of connexin43 was stronger than Gap27 to its corresponding loop EL1 (dissociation rate constant Kd 0.061 ± 0.004 vs. 0.0043 ± 0.0001 s-1, mean ± SD). Mitochondria from IPC hearts showed slightly but significantly reduced respiratory control ratio (RCR). In vitro addition of Gap peptides did not significantly alter respiration. If transient hemichannel activity is part of the IPC triggering event, inhibition of IPC triggering stimuli might limit the use of cardioprotective Gap peptides.


Subject(s)
Connexin 43 , Ischemic Preconditioning, Myocardial , Animals , Connexin 43/metabolism , Heart , Infarction , Ischemia , Male , Peptides/pharmacology , Rats
2.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681669

ABSTRACT

Aging attenuates cardiac tolerance to ischemia/reperfusion (I/R) associated with defects in protective cell signaling, however, the onset of this phenotype has not been completely investigated. This study aimed to compare changes in response to I/R and the effects of remote ischemic preconditioning (RIPC) in the hearts of younger adult (3 months) and mature adult (6 months) male Wistar rats, with changes in selected proteins of protective signaling. Langendorff-perfused hearts were exposed to 30 min I/120 min R without or with prior three cycles of RIPC (pressure cuff inflation/deflation on the hind limb). Infarct size (IS), incidence of ventricular arrhythmias and recovery of contractile function (LVDP) served as the end points. In both age groups, left ventricular tissue samples were collected prior to ischemia (baseline) and after I/R, in non-RIPC controls and in RIPC groups to detect selected pro-survival proteins (Western blot). Maturation did not affect post-ischemic recovery of heart function (Left Ventricular Developed Pressure, LVDP), however, it increased IS and arrhythmogenesis accompanied by decreased levels and activity of several pro-survival proteins and by higher levels of pro-apoptotic proteins in the hearts of elder animals. RIPC reduced the occurrence of reperfusion-induced ventricular arrhythmias, IS and contractile dysfunction in younger animals, and this was preserved in the mature adults. RIPC did not increase phosphorylated protein kinase B (p-Akt)/total Akt ratio, endothelial nitric oxide synthase (eNOS) and protein kinase Cε (PKCε) prior to ischemia but only after I/R, while phosphorylated glycogen synthase kinase-3ß (GSK3ß) was increased (inactivated) before and after ischemia in both age groups coupled with decreased levels of pro-apoptotic markers. We assume that resistance of rat heart to I/R injury starts to already decline during maturation, and that RIPC may represent a clinically relevant cardioprotective intervention in the elder population.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Aging , Animals , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Hemodynamics , Male , Myocardial Reperfusion Injury/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
3.
J Cell Mol Med ; 24(12): 6510-6522, 2020 06.
Article in English | MEDLINE | ID: mdl-32383522

ABSTRACT

Redox signalling in mitochondria plays an important role in myocardial ischaemia/reperfusion (I/R) injury and in cardioprotection. Reactive oxygen and nitrogen species (ROS/RNS) modify cellular structures and functions by means of covalent changes in proteins including among others S-nitros(yl)ation by nitric oxide (NO) and its derivatives, and S-sulphydration by hydrogen sulphide (H2 S). Many enzymes are involved in the mitochondrial formation and handling of ROS, NO and H2 S under physiological and pathological conditions. In particular, the balance between formation and removal of reactive species is impaired during I/R favouring their accumulation. Therefore, various interventions aimed at decreasing mitochondrial ROS accumulation have been developed and have shown cardioprotective effects in experimental settings. However, ROS, NO and H2 S play also a role in endogenous cardioprotection, as in the case of ischaemic pre-conditioning, so that preventing their increase might hamper self-defence mechanisms. The aim of the present review was to provide a critical analysis of formation and role of reactive species, NO and H2 S in mitochondria, with a special emphasis on mechanisms of injury and protection that determine the fate of hearts subjected to I/R. The elucidation of the signalling pathways of ROS, NO and H2 S is likely to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent I/R injury.


Subject(s)
Cardiotonic Agents/therapeutic use , Hydrogen Sulfide/metabolism , Mitochondria, Heart/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans
4.
Can J Physiol Pharmacol ; 98(3): 139-146, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31518503

ABSTRACT

Uric acid is a purine degradation product but also an important antioxidant and reactive oxygen species (ROS) scavenger. Experimental settings that mimic myocardial ischemia-reperfusion have not included uric acid despite that it is always present in human extracellular fluid and plasma. We hypothesized that uric acid has an important role in myocardial ROS scavenging. Here, we tested the cardiac response to uric acid on infarct size following ischemia-reperfusion with and without exacerbated oxidative stress due to acute pressure overload and during preconditioning. We also examined mitochondrial respiration and ROS-induced mitochondrial permeability transition pore opening. Under exacerbated ROS stress induced by high-pressure perfusion, uric acid lowered oxidative stress and reduced infarct size. In contrast, uric acid blocked cardioprotection induced by ischemic preconditioning. However, this effect was reversed by probenecid, an inhibitor of cellular uptake of uric acid. In accordance, in intact cardiomyocytes, extracellular uric acid reduced the susceptibility of mitochondria towards opening of the permeability transition pore, suggesting that uric acid may prevent ischemia-reperfusion injury due to scavenging of maladaptive ROS. Moreover, as uric acid also scavenges adaptive ROS, this may interfere with preconditioning. Altogether, uric acid might be a confounder when translating preclinical experimental results into clinical treatment.


Subject(s)
Antioxidants/pharmacology , Mitochondria, Heart/drug effects , Myocardial Ischemia/drug therapy , Myocardial Reperfusion Injury/drug therapy , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Uric Acid/pharmacology , Animals , Humans , Ischemic Preconditioning, Myocardial , Male , Mitochondria, Heart/pathology , Mitochondrial Permeability Transition Pore/metabolism , Myocardial Ischemia/etiology , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Rats , Rats, Wistar
5.
BMC Cardiovasc Disord ; 17(1): 79, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28292262

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction is closely associated with diastolic dysfunction and related to obesity and female sex. We investigated whether adiponectin, an adipocyte-secreted protein hormone with cardioprotective effects, was associated with indices of diastolic dysfunction, and whether the association was sex dependent. METHODS: We conducted a cross-sectional study on 1165 women and 896 men without diabetes. We stratified the multivariable adjusted logistic regression analyses and the fractional polynomial regression analyses according to sex, with echocardiographic markers of diastolic dysfunction as dependent variables, and adiponectin as the independent variable of interest. RESULTS: Decreased adiponectin was associated with higher odds of average tissue Doppler e' < 9 in women (odds ratio [OR] 1.17 per 1 µg/mL adiponectin decrease, 95% confidence interval [CI] 1.04-1.30), but not in men (p for interaction with sex 0.04). Women, but not men, had higher odds of E/e' ratio ≥ 8 with lower adiponectin (OR 1.12 per 1 µg/mL adiponectin decrease, 95% CI 1.02-1.24, p for interaction with sex 0.04). Adiponectin in the lower sex-specific tertile was associated with increased odds of concentric left ventricular hypertrophy in women (OR 2.44, 95% CI 1.03-5.77), but with decreased odds in men (OR 0.32, 95% CI 0.11-0.88, p for interaction with sex 0.002), and decreased odds of eccentric hypertrophy in men only (OR 0.53, 95% CI 0.33-0.88, p for interaction with sex 0.02). Adiponectin in the lower sex-specific tertile was associated with moderately enlarged left atria in women only (OR 1.43, 95% CI 1.01-2.03, p for interaction with sex 0.04). Finally, adiponectin had a non-linear relationship with left ventricular mass in women only, with exponentially increasing left ventricular mass with lower adiponectin levels (p for interaction with sex 0.01). CONCLUSIONS: Low adiponectin was associated with higher odds of indices of diastolic dysfunction in women, but lower odds of indices of diastolic dysfunction in men. Lower adiponectin was associated with increased left ventricular mass in women only.


Subject(s)
Adiponectin/blood , Heart Failure, Diastolic/blood , Heart Ventricles/physiopathology , Risk Assessment , Ventricular Function, Left/physiology , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Sectional Studies , Echocardiography, Doppler , Female , Heart Failure, Diastolic/epidemiology , Heart Failure, Diastolic/physiopathology , Heart Ventricles/diagnostic imaging , Humans , Incidence , Male , Middle Aged , Norway/epidemiology , Prognosis , Prospective Studies , Risk Factors , Sex Factors , Stroke Volume/physiology , Survival Rate/trends , Ventricular Remodeling
6.
Eur Heart J ; 37(23): 1789-98, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27055812

ABSTRACT

Despite improvements in modern cardiovascular therapy, the morbidity and mortality of ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and worldwide. Patients with IHD may benefit from therapies that would accelerate natural processes of postnatal collateral vessel formation and/or muscle regeneration. Here, we discuss the use of cells in the context of heart repair, and the most relevant results and current limitations from clinical trials using cell-based therapies to treat IHD and HF. We identify and discuss promising potential new therapeutic strategies that include ex vivo cell-mediated gene therapy, the use of biomaterials and cell-free therapies aimed at increasing the success rates of therapy for IHD and HF. The overall aim of this Position Paper of the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to improve the therapeutic application of cell-based therapies for cardiac regeneration and repair.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Heart Failure/therapy , Heart/physiology , Myocardial Ischemia/therapy , Cell Tracking/methods , Clinical Trials as Topic , Data Accuracy , Ethics, Medical , Heart Failure/physiopathology , Humans , Myocardial Ischemia/physiopathology , Patient Safety , Patient Selection , Regeneration/physiology , Stem Cell Transplantation/methods , Stroke Volume/physiology , Treatment Outcome
7.
Basic Res Cardiol ; 111(4): 42, 2016 07.
Article in English | MEDLINE | ID: mdl-27164906

ABSTRACT

Acute myocardial infarction (AMI) causes sterile inflammation, which exacerbates tissue injury. Elevated levels of circulating mitochondrial DNA (mtDNA) have been associated with AMI. We hypothesized that mtDNA triggers an innate immune response via TLR9 and NF-κB activation, causing cardiomyocyte injury. Murine cardiomyocytes express TLR9 mRNA and protein and were able to internalize fluorescently labeled mouse mtDNA. Incubation of human embryonic kidney cells with serum from AMI patients containing naturally elevated levels of mtDNA induced TLR9-dependent NF-κB activity. This effect was mimicked by isolated mtDNA. mtDNA activated NF-κB in reporter mice both in vivo and in isolated cardiomyocytes. Moreover, incubation of isolated cardiomyocytes with mtDNA induced cell death after 4 and 24 h. Laser confocal microscopy showed that incubation of cardiomyocytes with mtDNA accelerated mitochondrial depolarization induced by reactive oxygen species. In contrast to mtDNA, isolated total DNA did not activate NF-κB nor induce cell death. In conclusion, mtDNA can induce TLR9-dependent NF-κB activation in reporter cells and activate NF-κB in cardiomyocytes. In cardiomyocytes, mtDNA causes mitochondrial dysfunction and death. Endogenous mtDNA in the extracellular space is a danger signal with direct detrimental effects on cardiomyocytes.


Subject(s)
DNA, Mitochondrial/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Toll-Like Receptor 9/metabolism , Animals , Cell Death/physiology , Female , Humans , Immunoblotting , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Myocardial Infarction/metabolism , Polymerase Chain Reaction
8.
BMC Cardiovasc Disord ; 16: 85, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27165776

ABSTRACT

BACKGROUND: Elevated uric acid (UA) is associated with the presence of the metabolic syndrome (MetS). In a prospective cohort study, we assessed whether baseline and longitudinal change in UA were risk factors for development of MetS and its individual components. METHODS: We included 3087 women and 2996 men who had UA measured in the population based Tromsø Study 1994-95. The participants were stratified according to body mass index (BMI). Endpoints were MetS and each component of the syndrome after 7 years, according to the revised National Cholesterol Education Program's Adult Treatment Panel III (NCEP-ATP III) definition. RESULTS: Multiple logistic regression analyses showed that higher baseline UA was associated with higher odds of developing elevated blood pressure in overweight subjects (BMI ≥ 25 kg/m(2), odds ratio [OR] per 59 µmol/L UA increase 1.44, 95 % confidence interval [CI] = 1.17-1.77, P = 0.001), but not in normal-weight subjects (BMI < 25 kg/m(2), P for interaction = 0.04). Overweight also modified the association between baseline UA and the development of elevated fasting glucose (P for interaction = 0.01). UA was a predictor of MetS in all subjects (OR per 59 µmol/L UA increase 1.29, 95 % CI 1.18-1.41, P < 0.001). Furthermore, longitudinal UA change was independently associated with the development of MetS in all subjects (OR per 59 µmol/L UA increase over 7 years 1.28, 95 % CI 1.16-1.42, P < 0.001). CONCLUSION: Increased levels of baseline UA independently predicted development of elevated blood pressure and higher fasting glycemia in the overweight, but not the normal-weight subjects. Baseline UA and longitudinal increase in UA over 7 years was associated with the development of MetS in all subjects. Whether increased UA should be treated differently in normal-weight and overweight persons needs further study.


Subject(s)
Hyperuricemia/epidemiology , Metabolic Syndrome/epidemiology , Obesity/epidemiology , Uric Acid/blood , Aged , Biomarkers/blood , Blood Glucose/analysis , Blood Pressure , Body Mass Index , Chi-Square Distribution , Female , Humans , Hyperglycemia/blood , Hyperglycemia/epidemiology , Hypertension/epidemiology , Hypertension/physiopathology , Hyperuricemia/blood , Hyperuricemia/diagnosis , Linear Models , Logistic Models , Longitudinal Studies , Male , Metabolic Syndrome/blood , Metabolic Syndrome/diagnosis , Metabolic Syndrome/physiopathology , Middle Aged , Multivariate Analysis , Norway/epidemiology , Obesity/diagnosis , Obesity/physiopathology , Odds Ratio , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , Up-Regulation
9.
Tidsskr Nor Laegeforen ; 137(22)2017 11 28.
Article in Norwegian | MEDLINE | ID: mdl-29181949
10.
Am J Physiol Cell Physiol ; 300(4): C783-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21209360

ABSTRACT

The therapeutic utility of endothelial progenitor cells (EPCs) in cardiovascular disease is potentially hampered by their low numbers in the circulation, impaired functional activity, and inhibitory factors in the recipient. These obstacles can possibly be circumvented by the use of proangiogenic cytokines and peptides. We sought to examine the effect of the endogenous vasoactive peptide adrenomedullin (AM) on the angiogenic potential of late outgrowth EPCs and their release of proangiogenic and proinflammatory cytokines/chemokines. Human peripheral blood mononuclear cells were cultured until the appearance of typical late outgrowth EPC colonies. The effect of AM on EPC proliferation was assessed using a colorimetric MTS proliferation assay while differentiation and formation of tubular structures in an EPC/fibroblast coculture or matrigel assay was used to assess the angiogenic potential of the cells. Finally, the release and mRNA transcripts of cytokines/chemokines were quantified in stimulated vs. nonstimulated EPCs using real-time PCR and a bead-based multiplex assay. The cultured EPCs possessed an endothelial phenotype and expressed the AM receptor (calcitonin receptor-like receptor/receptor activity modifying protein-2). AM stimulation induced proliferation of EPCs compared with controls (P < 0.05). Furthermore, AM produced a 36% and 80% increase in the formation of tubular networks in the EPC/fibroblast coculture and matrigel assay, respectively (P < 0.05). These effects seemed to be mediated through the phosphatidylinositol 3-kinase/Akt signaling pathway. AM did not seem to significantly influence the release or production of IL-6, IL-8, VEGF, stromal cell-derived factor 1, or the expression of CXCR-4 or VEGF receptor 2. In conclusion, adrenomedullin augmented the growth and angiogenic properties of late outgrowth EPCs, but did not influence their paracrine properties.


Subject(s)
Adrenomedullin/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Neovascularization, Physiologic/drug effects , Stem Cells/drug effects , Stem Cells/physiology , Cell Proliferation/drug effects , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Neovascularization, Physiologic/physiology , Stem Cells/cytology , Vascular Endothelial Growth Factor A/metabolism
11.
Cell Physiol Biochem ; 28(1): 103-14, 2011.
Article in English | MEDLINE | ID: mdl-21865853

ABSTRACT

AIM: Gap junction intercellular communication (GJIC) and hemichannel permeability may have important roles during an ischemic insult. Our aim was to evaluate the effect of ischemia on gap junction channels and hemichannels. METHODS: We used neonatal rat heart myofibroblasts and simulated ischemia with a HEPES buffer with high potassium, low pH, absence of glucose, and oxygen tension was reduced by dithionite. Microinjection, western blot, immunofluorescence, cell viability and dye uptake were used to evaluate the effects induced by dithionite. Isolated perfused rat hearts were used to analyse infarct size. RESULTS: Short period with simulated ischemia reduced the ability to transfer a dye between neighbouring cells, which indicated reduced GJIC. Prolonged exposure to simulated ischemia caused opening of hemichannels, and cell death was apparent while gap junction channels remained closed. Connexin 43 became partially dephosphorylated and the total amount decreased during simulated ischemia. We were not able to detect the alternative hemichannel-forming protein, Pannexin 1, in these cells. The potential importance of Connexin 43 or Pannexin 1 hemichannels in ischemia-induced infarct in the intact heart was studied by perfusion of the heart in the presence of peptides that block one or the other type of hemichannels. The connexin-derived peptide, Gap26, significantly reduced the infract/risk zone ratio (control 48.7±4.2% and Gap26 19.4±4.1%, p<0.001), while the pannexin-derived peptide, (10)Panx1, did not change infarct/risk ratio. CONCLUSION: Connexin 43 is most likely responsible for both closure of gap junction channels and opening of hemichannels during simulated ischemia in neonatal rat heart myofibroblasts. Opening of connexin 43 hemichannels during ischemia-reperfusion seems to be an important mechanism for ischemia-reperfusion injury in the heart. By preventing the opening of these channels during early ischemia-reperfusion the infarct size becomes significantly reduced.


Subject(s)
Gap Junctions/metabolism , Ischemia/metabolism , Animals , Animals, Newborn , Cell Communication , Cells, Cultured , Connexin 43/metabolism , Connexin 43/physiology , Connexins/metabolism , Dithionite/pharmacology , Female , Ischemia/physiopathology , Myofibroblasts/metabolism , Myofibroblasts/physiology , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Rats , Rats, Wistar
12.
Front Physiol ; 12: 663819, 2021.
Article in English | MEDLINE | ID: mdl-34349662

ABSTRACT

Angiotensin-converting enzyme 2 (ACE 2) in the heart including its sex dependency in the hypertensive heart, has not been much studied compared to ACE. In the present study, we used the Dahl salt-sensitive rat exposed to fructose and salt to model a hypertensive phenotype in males, females, and ovariectomized females. Blood pressure was measured by the tale-cuff technique in the conscious state. Expression of RAS-related genes ACE, ACE2, angiotensin II receptor type 1, Mas1, and CMA1 in the heart were quantified. The results revealed small but significant differences between male and female groups. The main results indicate the presence of a male preponderance for an increase in ACE and ACE2 gene expression. The results are in accordance with the role of androgens or male chromosomal complement in controlling the expression of the two ACE genes.

13.
Cardiovasc Res ; 117(2): 367-385, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32484892

ABSTRACT

Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.


Subject(s)
Health Status Disparities , Myocardial Ischemia/epidemiology , Translational Research, Biomedical , Animals , Comorbidity , Disease Models, Animal , Female , Humans , Male , Myocardial Ischemia/diagnosis , Myocardial Ischemia/physiopathology , Risk Assessment , Risk Factors , Sex Characteristics , Sex Factors , Species Specificity
15.
Scand Cardiovasc J ; 44(3): 183-90, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20233134

ABSTRACT

OBJECTIVES: To investigate the protective effects of adenosine against hypoxic injury at hypothermia; both magnitude and mechanisms. DESIGN: Receptor versus non-receptor dependent mechanisms in cardioprotection by adenosine were examined in guinea pig papillary muscles exposed to glucose free hypoxia at 24 degrees C. Contractile force amplitude (CFA) and action potential duration (APD) during increasing concentrations of adenosine at 37 degrees C, 30 degrees C and 24 degrees C normoxia were also examined. RESULTS: CFA was significantly improved after adenosine treatment during hypothermic hypoxia compared to control (80.7+/-17.4% vs 40.5+/-10.7%, p<0.001). Adenosine receptor antagonist SPT did not antagonize (64.6+/-21.1%), and adenosine receptor agonists (APNEA+NECA) could not mimic the cardioprotection (53.8+/-9.3%). MitoK(Ca) blocker paxilline antagonized the cardioprotection (40.0+/-7.7%). During normoxic conditions hypothermia-induced increase in CFA was significantly decreased by adenosine (0.12-12 mM) whereas the increase in action potential duration was potentiated. CONCLUSION: Adenosine (1.2 mM) had marked cardioprotective effect in hypothermic substrate free hypoxia. Possible mechanisms are non-receptor dependent and related to mitoK(Ca) channels. The cardiodepressive effect at hypothermia may contribute to cardioplegia.


Subject(s)
Adenosine/pharmacology , Heart Arrest, Induced/methods , Hypothermia, Induced , Myocardial Contraction/drug effects , Papillary Muscles/drug effects , Potassium Compounds/pharmacology , Action Potentials , Animals , Cell Hypoxia , Dose-Response Relationship, Drug , Female , Guinea Pigs , In Vitro Techniques , Indoles/pharmacology , Male , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/metabolism , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/metabolism , Time Factors
16.
Br J Pharmacol ; 177(23): 5270-5286, 2020 12.
Article in English | MEDLINE | ID: mdl-31863453

ABSTRACT

Translation of cardioprotective interventions aimed at reducing myocardial injury during ischaemia-reperfusion from experimental studies to clinical practice is an important yet unmet need in cardiovascular medicine. One particular challenge facing translation is the existence of demographic and clinical factors that influence the pathophysiology of ischaemia-reperfusion injury of the heart and the effects of treatments aimed at preventing it. Among these factors, age and sex are prominent and have a recognised role in the susceptibility and outcome of ischaemic heart disease. Remarkably, some of the most powerful cardioprotective strategies proven to be effective in young animals become ineffective during ageing. This article reviews the mechanisms and implications of the modulatory effects of ageing and sex on myocardial ischaemia-reperfusion injury and their potential effects on cardioprotective interventions. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.


Subject(s)
Cardiovascular Agents , Myocardial Ischemia , Myocardial Reperfusion Injury , Aging , Animals , Heart , Myocardial Reperfusion Injury/prevention & control
17.
Cryobiology ; 59(3): 297-301, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19723518

ABSTRACT

Hypothermia-rewarming of the heart results in contractile dysfunction under in vitro as well as in vivo conditions. Increase in reactive oxygen species (ROS), lipid peroxidation and calcium overload are proposed mechanisms. In the first protocol of this study, the effect of putative phospholipase and calcium channel modulator mepacrine during deep hypothermia (4 h 14 degrees C) plus rewarming was tested in an isolated perfused rat heart model previously reported not to involve increase in lipid peroxides. Contractile function was measured under isovolumetric conditions using an intra-ventricular balloon connected to a transducer and recording system. Mepacrine completely reversed hypothermia-rewarming induced contractile failure in this model (LV dP/dt(max): 3236+/-517 vs. 1058+/-185 mmHg/s in untreated hearts). In the second part of the study, lipid peroxidation of the heart was examined in vivo in anesthetized rats subjected to 4h of deep hypothermia followed by rewarming. In this model recovery of heart function judged by cardiac output is decreased whereas blood pressure and heart rate recover fully. Peroxy conjugated diene isomers of unsaturated fatty acids were measured in heart phospholipids. The composition of the non-esterified fatty acids and the phospholipid fatty acid pool was examined in order to reveal signs of membrane remodeling. The results demonstrated no significant changes in phospholipid peroxidation after rewarming (91.07+/-5.23 vs. 88.63+/-7.73 nmol/g dry wt. in control). There was significant relative reduction in the content of arachidonic acid in the phospholipid fraction (29.55+/-1.65 vs. 24.76+/-1.48%). There was marked decrease in non-esterified fatty acids in myocardial tissue (1992+/-291 vs. 1069+/-189 nmol/g dry wt.), but a significant relative increase in arachidonic acid (20:4) in this fraction (3.46+/-0.42 vs. 4.99+/-0.30%). In conclusion, rewarming from deep hypothermia is not associated with increased phospholipid peroxidation. There is, however, a significant remodeling of the phospholipid fraction of myocardial lipids in vivo probably as a result of receptor or calcium stimulated phospholipase activity. Calcium or calcium stimulated phospholipase activity could contribute to posthypothermic contractile dysfunction.


Subject(s)
Hypothermia, Induced/adverse effects , Quinacrine/pharmacology , Rewarming/adverse effects , Animals , Arachidonic Acid/metabolism , Blood Pressure/drug effects , Cardiac Output , Heart/physiology , Heart Rate/drug effects , In Vitro Techniques , Lipid Peroxidation/drug effects , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Phospholipids/metabolism , Rats , Rats, Wistar
18.
Sci Rep ; 9(1): 10513, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324844

ABSTRACT

Several epidemiological studies have pointed at serum uric acid (SUA) as an independent risk factor for mortality, diabetes, hypertension, cardiovascular and kidney disease; however, no clear pathogenic pathway is established. Uric acid (UA) crystals show pro-inflammatory properties and can thus create or contribute to the state of chronic low-grade inflammation, a widely accepted pathogenic mechanism in several of the above-mentioned pathologies. On the other hand, soluble uric acid possesses antioxidant properties that might attenuate inflammatory responses. We aimed to explore the net effects of experimentally rising SUA in human whole blood cultures on several mediators of inflammation. Production of TNF-α, IL-1ß, IL-1RA, MCP-1 and IL-8 was assessed upon addition of 200 µM UA, 500 µM UA or monosodium urate (MSU) crystals in the presence or absence of 5 ng/ml lipopolysaccharide (LPS). RT-qPCR and multiplex bead based immunoassay were used to measure mRNA expression and cytokine release at 2 and 4 h of culture, respectively. 14C labeled UA was used to assess intracellular uptake of UA. We show that crystallized, but not soluble, UA induces production of pro-inflammatory mediators in human whole blood. Soluble UA is internalized in blood cells but does not potentiate or reduce LPS-induced release of cytokines.


Subject(s)
Blood Cells/drug effects , Inflammation/blood , Uric Acid/pharmacology , Blood Cells/metabolism , Blood Culture , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Crystallization , Female , Humans , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-8/biosynthesis , Interleukin-8/genetics , Lipopolysaccharides/pharmacology , Male , RNA, Messenger/biosynthesis , Solubility , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Uric Acid/chemistry
19.
Cardiovasc Res ; 115(3): 488-500, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30657875

ABSTRACT

Morbidity and mortality from ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and are increasing worldwide. Patients with IHD or HF might benefit from novel therapeutic strategies, such as cell-based therapies. We recently discussed the therapeutic potential of cell-based therapies and provided recommendations on how to improve the therapeutic translation of these novel strategies for effective cardiac regeneration and repair. Despite major advances in optimizing these strategies with respect to cell source and delivery method, the clinical outcome of cell-based therapy remains unsatisfactory. Major obstacles are the low engraftment and survival rate of transplanted cells in the harmful microenvironment of the host tissue, and the paucity or even lack of endogenous cells with repair capacity. Therefore, new ways of delivering cells and their derivatives are required in order to empower cell-based cardiac repair and regeneration in patients with IHD or HF. Strategies using tissue engineering (TE) combine cells with matrix materials to enhance cell retention or cell delivery in the transplanted area, and have recently received much attention for this purpose. Here, we summarize knowledge on novel approaches emerging from the TE scenario. In particular, we will discuss how combinations of cell/bio-materials (e.g. hydrogels, cell sheets, prefabricated matrices, microspheres, and injectable matrices) combinations might enhance cell retention or cell delivery in the transplantation areas, thereby increase the success rate of cell therapies for IHD and HF. We will not focus on the use of classical engineering approaches, employing fully synthetic materials, because of their unsatisfactory material properties which render them not clinically applicable. The overall aim of this Position Paper from the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to proceed in research with these novel TE strategies combined with cell-based therapies to boost cardiac repair in the clinical settings of IHD and HF.


Subject(s)
Biomedical Research/standards , Cardiology/standards , Heart Failure/surgery , Myocardial Ischemia/surgery , Myocardium/pathology , Regeneration , Stem Cell Transplantation/standards , Tissue Engineering/standards , Consensus , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Recovery of Function , Stem Cell Transplantation/adverse effects , Treatment Outcome
20.
J Pineal Res ; 45(4): 449-58, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18691357

ABSTRACT

Melatonin has potent cardioprotective properties. These actions have been attributed to its free radical scavenging and anti-oxidant actions, but may also be receptor mediated. Melatonin also exerts powerful anti-adrenergic actions based on its effects on contractility of isolated papillary muscles. The aims of this study were to determine whether melatonin also has anti-adrenergic effects on the isolated perfused rat heart, to determine the mechanism thereof and to establish whether these actions contribute to protection of the heart during ischaemia/reperfusion. The results showed that melatonin (50 microM) caused a significant reduction in both isoproterenol (10(-7) M) and forskolin (10(-6) M) induced cAMP production and that both these responses were melatonin receptor dependent, since the blocker, luzindole (5 x 10(-6) M) abolished this effect. Nitric oxide (NO), as well as guanylyl cyclase are involved, as L-NAME (50 microM), an NO synthase inhibitor and ODQ (20 microM), a guanylyl cyclase inhibitor, significantly counteracted the effects of melatonin. Protein kinase C (PKC), as indicated by the use of the inhibitor bisindolylmaleimide (50 microM), also play a role in melatonin's anti-adrenergic actions. These actions of melatonin are involved in its cardioprotection: simultaneous administration of L-NAME or ODQ with melatonin, before and after 35 min regional ischaemia, completely abolished its cardioprotection. PKC, on the other hand, had no effect on the melatonin-induced reduction in infarct size. Cardioprotection by melatonin was associated with a significant activation of PKB/Akt and attenuated activation of the pro-apoptotic kinase, p38MAPK during early reperfusion. In summary, the results show that melatonin-induced cardioprotection may be receptor dependent, and that its anti-adrenergic actions, mediated by NOS and guanylyl cyclase activation, are important contributors.


Subject(s)
Adrenergic Antagonists/pharmacology , Cyclic AMP/metabolism , Melatonin/pharmacology , Myocardial Reperfusion Injury/prevention & control , Receptors, Melatonin/metabolism , Animals , Colforsin/pharmacology , Guanylate Cyclase/metabolism , Heart/drug effects , Heart/physiology , Indoles/pharmacology , Isoproterenol/pharmacology , Male , Maleimides/pharmacology , Melatonin/agonists , Myocardial Infarction/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/physiology , Oxadiazoles/pharmacology , Propranolol/pharmacology , Protein Kinase C/metabolism , Quinoxalines/pharmacology , Rats , Rats, Wistar , Tryptamines/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL