Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 111(6): 70, 2016 11.
Article in English | MEDLINE | ID: mdl-27766474

ABSTRACT

To commemorate the auspicious occasion of the 30th anniversary of IPC, leading pioneers in the field of cardioprotection gathered in Barcelona in May 2016 to review and discuss the history of IPC, its evolution to IPost and RIC, myocardial reperfusion injury as a therapeutic target, and future targets and strategies for cardioprotection. This article provides an overview of the major topics discussed at this special meeting and underscores the huge importance and impact, the discovery of IPC has made in the field of cardiovascular research.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury , Animals , Humans
2.
J Physiol ; 591(2): 461-73, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23090948

ABSTRACT

Hypoxia-reoxygenation induces loss of endothelial barrier function and oedema formation, which presents a major impediment for recovery of the organ. The integrity of the endothelial barrier is highly dependent on its contractile machinery and actin dynamics, which are precisely regulated by Rho GTPases. Perturbed activities of these Rho-GTPases under hypoxia-reoxygenation lead to derangement of the actin cytoskeleton and therefore may affect the integrity of the endothelial barrier. The aim of the present study was to analyse the role of these GTPases in regulating endothelial barrier function during hypoxia-reoxygenation in cultured porcine aortic endothelial cells and isolated perfused rat hearts. Hypoxia-reoxygenation induced an increase in albumin permeability of endothelial monolayers accompanied by an activation of the endothelial contractile machinery, derangement of the actin cytoskeleton and loss of VE-cadherin from cellular junctions. Inhibition of contractile activation with ML-7 partially protected against hypoxia-reoxygenation-induced hyperpermeability. Likewise, reoxygenation caused an increase in RhoA and a reduction in Rac1 activity accompanied by enhanced stress fibre formation and loss of peripheral actin. Inhibition of RhoA/rho kinase (Rock) signalling with RhoA or Rock inhibitors led to a complete depolymerisation and derangement of the actin cytoskeleton and worsened hypoxia-reoxygenation-induced hyperpermeability. Activation of Rac1 using a cAMP analogue, 8-CPT-O-Me-cAMP, which specifically activates Epac/Rap1 signalling, restored peripheral localisation of actin and VE-cadherin at cellular junctions and abrogated reoxygenation-induced hyperpermeability. Similar results were reproduced in isolated saline-perfused rat hearts. These data show that activation of Rac1 but not the inhibition of RhoA preserves endothelial integrity against reoxygenation-induced loss of barrier function.


Subject(s)
Endothelial Cells/metabolism , Muscle, Smooth, Vascular/physiology , Myosin-Light-Chain Kinase/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD/metabolism , Aorta/cytology , Aorta/physiology , Cadherins/metabolism , Calcium/metabolism , Cell Hypoxia , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Permeability , Rats , Signal Transduction , Stress Fibers/metabolism , Swine , Vasoconstriction , rho-Associated Kinases/metabolism
3.
Cardiovasc Res ; 94(2): 168-80, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22499772

ABSTRACT

Reperfusion may induce additional cell death in patients with acute myocardial infarction receiving primary angioplasty or thrombolysis. Altered intracellular Ca(2+) handling was initially considered an essential mechanism of reperfusion-induced cardiomyocyte death. However, more recent studies have demonstrated the importance of Ca(2+)-independent mechanisms that converge on mitochondrial permeability transition (MPT) and are shared by cardiomyocytes and other cell types. This article analyses the importance of Ca(2+)-dependent cell death in light of these new observations. Altered Ca(2+) handling includes increased cytosolic Ca(2+) levels, leading to activation of calpain-mediated proteolysis and sarcoplasmic reticulum-driven oscillations; this can induce hypercontracture, but also MPT due to the privileged Ca(2+) transfer between sarcoplasmic reticulum and mitochondria through cytosolic Ca(2+) microdomains. In the opposite direction, permeability transition can worsen altered Ca(2+) handling and favour hypercontracture. Ca(2+) appears to play an important role in cell death during the initial minutes of reperfusion, particularly after brief periods of ischaemia. Developing effective and safe treatments to prevent Ca(2+)-mediated cardiomyocyte death in patients with transient ischaemia, by targeting Ca(2+) influx, intracellular Ca(2+) handling, or Ca(2+)-induced cell death effectors, is an unmet challenge with important therapeutic implications and large potential clinical impact.


Subject(s)
Calcium/metabolism , Mitochondria, Heart/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion/adverse effects , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Calpain/metabolism , Cell Death , Humans , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Sarcoplasmic Reticulum/metabolism
5.
J Mol Cell Cardiol ; 52(5): 962-70, 2012 May.
Article in English | MEDLINE | ID: mdl-22266063

ABSTRACT

ATP can differentially affect the micro- and macrovascular endothelial barrier. It has been shown that it can both increase and/or decrease macromolecule permeability of microvascular endothelial cells and microvessels, in vivo. We hypothesised that the barrier stabilising effect is mediated by ATP itself via P2 receptors, while barrier-disrupting effect is mediated by its metabolite adenosine via adenosine receptors. The effects of ATP, ADP, AMP and adenosine on barrier function were studied in cultured rat coronary microvascular endothelial monolayers (RCEC) in vitro, as well as in rat mesentery vessels, and in rat hearts in vivo. ATP and ADP showed a biphasic effect on permeability of RCEC monolayers with a reduction followed by a later increase in albumin permeability. The permeability decreasing effect of ATP was enhanced by ecto-nucleotidase inhibitor ARL67156 while permeability increasing effect was enhanced by apyrase, an extracellular ecto-nucleotidase. Moreover, the permeability increasing effect was abrogated by adenosine receptor antagonists, 8-phenyltheophylline (8-PT) and DMPX. Adenosine and adenosine receptor agonists 5'-(N-ethylcarboxamido)-adenosine (NECA), CGS21680, and R-PIA enhanced albumin permeability which was antagonised by 8-PT, A(1), and A(2) but not by A(3) receptor antagonists. Likewise, immunofluorescence microscopy of VE-cadherin and actin showed that NECA induces a disturbance of intercellular junctions. Pre-incubation of ATP antagonised the effects of NECA on permeability, actin cytoskeleton and intercellular junctions. Similar effects of the applied substances were observed in rat mesentery artery by determining the vascular leakage using intravital microscopy as well as in rat hearts by assessing myocardial water contents in vivo. In conclusion, the study demonstrates that in RCEC, ATP, ADP, and its metabolite adenosine play opposing roles on endothelial barrier function.


Subject(s)
Adenosine Triphosphate/pharmacology , Adenosine/pharmacology , Coronary Vessels/physiology , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Venules/physiology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Animals , Cadherins/metabolism , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Edema, Cardiac/pathology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Male , Myocardium , Permeability/drug effects , Protein Transport/drug effects , Purinergic P2 Receptor Antagonists/pharmacology , Rats , Rats, Wistar , Receptors, Purinergic P1/metabolism , Theophylline/analogs & derivatives , Theophylline/pharmacology , Venules/cytology , Venules/drug effects , Venules/metabolism
8.
Cardiovasc Res ; 92(2): 276-86, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21816966

ABSTRACT

AIMS: Intermedin (IMD) is a novel member of the calcitonin gene-related peptide family, which acts via calcitonin receptor-like receptors (CLRs), mediating activation of cAMP signalling. The main objective of the present study was to analyse the molecular mechanisms of the differential effects of IMD on the macromolecule permeability of endothelial cells of different vascular beds. METHODS AND RESULTS: Here we demonstrate that IMD increases permeability of rat coronary microvascular endothelial cells (RCECs) and reduces permeability of human umbilical vein endothelial cells (HUVECs) and rat aortic endothelial cells via CLRs and cAMP. Intermedin causes a derangement of the actin cytoskeleton accompanied by loss of vascular endothelial cadherin (VE-cadherin) in RCECs, while it causes a rearrangement of the actin cytoskeleton and VE-cadherin at cell-cell junctions in HUVECs. Intermedin inactivates the RhoA/Rho-kinase (Rock) pathway in both cell types; however, it inactivates Rac1 in RCECs but not in HUVECs. Inhibition and rescue experiments demonstrate that both RhoA and Rac1 are required for the RCEC barrier stability, while in HUVECs the inhibition of RhoA/Rock signalling does not interfere with basal permeability. CONCLUSION: The opposite effects of IMD on permeability of RCECs and HUVECs are due to differential regulation of actin cytoskeleton dynamics via RhoA and Rac1. Moreover, Rac1 activity is regulated by the RhoA/Rock pathway in RCECs but not in HUVECs.


Subject(s)
Actin Cytoskeleton/enzymology , Adrenomedullin/metabolism , Capillary Permeability , Coronary Vessels/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , Microvessels/enzymology , Neuropeptides/metabolism , Peptide Hormones/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Calcitonin Receptor-Like Protein/metabolism , Cells, Cultured , Coronary Vessels/cytology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Male , Microvessels/cytology , Rats , Rats, Wistar , Receptors, Calcitonin/metabolism , Time Factors , Transfection , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics
9.
J Cell Physiol ; 226(10): 2683-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792926

ABSTRACT

UNLABELLED: Transforming growth factor ß (TGFß) expression is induced in the myocardium during transition from compensated hypertrophy to heart failure. In cardiomyocytes, stimulation with TGFß results in restricted contractile function and enhanced apoptosis. Nitric oxide (NO) also induces apoptosis and influences cardiac function. Therefore, we wanted to know whether NO is causally involved in TGFß-induced apoptosis. In isolated ventricular cardiomyocytes of adult rat incubation with TGFß(1) increased NO release which was inhibited by NOS inhibitor ETU but not with iNOS inhibitor (1400 W) or nNOS inhibitor (TFA). In addition, TGFß-induced apoptosis was blocked with ETU and ODQ, but not with 1400 W or TFA. The consequent assumption that endothelial NOS is involved in TGFß-induced NO formation and apoptosis was supported by increased phosphorylation of eNOS at serine 1177 and by the fact that TGFß did not increase NO release in eNOS KO mice. Furthermore, TGFß-induced apoptosis, NO formation, SMAD binding activity and SMAD2 phosphorylation were blocked by a TGFß receptor antagonist, but only apoptosis and NO formation could be blocked with ETU. Expression of SMAD7 was increased after TGFß stimulation and blocked with TGFß receptor antagonist but not after blocking NO synthase with ETU. CONCLUSION: In cardiomyocytes TGFß-induced apoptosis is mediated via TGFß receptor activation that concomitantly activates SMAD transcription factors and the eNOS/NO/sGC pathway. Both of these pathways are needed for apoptosis induction by TGFß. This reveals a new pathway of cardiac NO release and identifies NO as a possible contributor to heart failure progression mediated by TGFß.


Subject(s)
Apoptosis/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Male , Myocytes, Cardiac/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Wistar , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
10.
Cardiovasc Res ; 87(2): 375-84, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20202976

ABSTRACT

AIMS: Activation of cAMP signalling abrogates thrombin-induced hyperpermeability. One of the mechanisms underlying this protective effect is the inactivation of endothelial contractile machinery, one of the major determinants of endothelial barrier function, mainly via the activation of myosin light chain phosphatase (MLCP). To date, the mechanisms of cAMP-mediated MLCP activation are only partially understood. Here the contribution of two cAMP effectors, PKA and Epac, in the regulation of endothelial contractile machinery and barrier function was studied. METHODS AND RESULTS: Endothelial contractile machinery and barrier function were analysed in cultured human umbilical vein endothelial cells (HUVEC). The cAMP analogues 8-CPT-cAMP and 6-Bnz-cAMP were used to activate Epac and PKA, respectively, and forskolin (FSK) was used to activate adenylyl cyclase. The cells were challenged by thrombin to inhibit MLCP via the RhoA/Rock pathway. Activation of either PKA or Epac partially blocked thrombin-induced hyperpermeability. Simultaneous activation of PKA and Epac had additive effects that were comparable to that of FSK. Activation of PKA but not Epac inhibited thrombin-induced phosphorylation of MLC and the MLCP regulatory subunit MYPT1, partly via inhibition of the RhoA/Rock pathway. FSK activated the MLCP catalytic subunit PP1 via dephosphorylation and dissociation of the PP1 inhibitory protein CPI-17. FSK blunted thrombin-induced CPI-17 phosphorylation, CPI-17/PP1 complex formation, and PP1 inactivation. Down-regulation of CPI-17 attenuated thrombin-induced hyperpermeability and abolished the antagonistic effect of the PKA activator, whereas the Epac activator retained its antagonistic effect. CONCLUSION: cAMP/PKA regulates the endothelial barrier via inhibition of the contractile machinery, mainly by the activation of MLCP via inhibition of CPI-17 and RhoA/Rock. The permeability-lowering effect of the cAMP/Epac pathway is independent of CPI-17.


Subject(s)
Capillary Permeability , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Endothelial Cells/enzymology , Myosin-Light-Chain Phosphatase/metabolism , Phosphoprotein Phosphatases/metabolism , Thrombin/metabolism , Adenylyl Cyclases/metabolism , Capillary Permeability/drug effects , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Enzyme Activation , Enzyme Activators/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Muscle Proteins , Myosin Light Chains/metabolism , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Phosphatase 1/metabolism , RNA Interference , Signal Transduction , Thionucleotides/pharmacology , Time Factors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
11.
Arterioscler Thromb Vasc Biol ; 30(6): 1237-45, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20339116

ABSTRACT

OBJECTIVE: Insulin is a key regulator of metabolism, but it also confers protective effects on the cardiovascular system. Here, we analyze the mechanism by which insulin stabilizes endothelial barrier function. METHODS AND RESULTS: Insulin reduced basal and antagonized tumor necrosis factor-alpha-induced macromolecule permeability of rat coronary microvascular endothelial monolayers. It also abolished reperfusion-induced vascular leakage in isolated-perfused rat hearts. Insulin induced dephosphorylation of the regulatory myosin light chains, as well as translocation of actin and vascular endothelial (VE)-cadherin to cell borders, indicating a reduction in contractile activation and stabilization of cell adhesion structures. These protective effects were blocked by genistein or Hydroxy-2-naphthalenylmethylphosphonic acid tris acetoxymethyl ester (HNMPA-[AM](3)), a pan-tyrosine-kinase or specific insulin-receptor-kinase inhibitor, respectively. Insulin stimulated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway and NO production, and it activated Rac1. Inhibition of PI3K/Akt abrogated Rac1 activation and insulin-induced barrier protection, whereas inhibition of the endothelial nitric oxide synthase/soluble guanylyl cyclase pathway partially inhibited them. Inhibition of Rac1 abrogated the assembly of actin at cell borders. Accordingly, it abolished the protective effect of insulin on barrier function of the cultured endothelial monolayer, as well as the intact coronary system of ischemic-reperfused hearts. CONCLUSIONS: Insulin stabilizes endothelial barrier via inactivation of the endothelial contractile machinery and enhancement of cell-cell adhesions. These effects are mediated via PI3K/Akt- and NO/cGMP-induced Rac1 activation.


Subject(s)
Capillary Permeability , Coronary Vessels/enzymology , Endothelial Cells/enzymology , Insulin/metabolism , Microvessels/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , rac1 GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Cyclic GMP/metabolism , Endothelial Cells/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/metabolism , Male , Microvessels/cytology , Microvessels/drug effects , Myosin Light Chains/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Rats, Wistar , Receptor, Insulin/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Soluble Guanylyl Cyclase , Time Factors , Tumor Necrosis Factor-alpha/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors
14.
J Cell Physiol ; 220(2): 515-23, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19415695

ABSTRACT

Heart failure development goes along with a transition from hypertrophic growth to apoptosis induction. In adult cardiomyocytes SMAD proteins are only activated under apoptotic, but not under hypertrophic conditions and are increased at the transition to heart failure. Therefore, SMADs could be candidates that turn the balance from hypertrophic growth to apoptosis resulting in heart failure development. To test this hypothesis we infected isolated rat ventricular cardiomyocytes with adenovirus encoding SMAD4 (AdSMAD4) and investigated the impact of SMAD4 overexpression on the development of apoptosis and hypertrophy under stimulation with phenylephrine (PE). Infection of cardiomyocytes with AdSMAD4 significantly enhanced SMAD-binding activity while apoptosis after 24 and 36 h infection did not rise. But when SMAD4 overexpressing cardiomyocytes were incubated with PE (10 microM), the number of apoptotic cells increased (Ctrl: 94.97 +/- 6.91%; PE: 102.48 +/- 4.78% vs. AdSMAD4 + PE: 118.64 +/- 3.28%). Furthermore expression of caspase 3 as well as bax/bcl2 ratio increased in SMAD4 overexpressing, PE-stimulated cardiomyocytes. In addition, the effects of SMAD4 overexpression on PE-induced hypertrophic growth were analyzed. Protein synthesis 36 h after AdSMAD4 infection was comparable to control cells, whereas the increase in protein synthesis stimulated by phyenylephrine was significantly reduced in SMAD4 overexpressing cells (134.28 +/- 10.02% vs. 100.57 +/- 8.86%). SMAD4 triggers the transition from hypertrophy to apoptosis in ventricular cardiomyocytes. Since SMADs are increased under several pathophysiological conditions in the heart, it can be assumed that it triggers apoptosis induction and therefore contributes to negative remodeling and heart failure progression.


Subject(s)
Apoptosis/physiology , Heart Ventricles/cytology , Hypertrophy/metabolism , Myocytes, Cardiac/metabolism , Smad4 Protein/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cells, Cultured , Heart Ventricles/metabolism , Hypertrophy/pathology , Male , Myocytes, Cardiac/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Wistar , Smad4 Protein/genetics , Transforming Growth Factor beta/metabolism
15.
Cardiovasc Res ; 83(2): 381-7, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19234300

ABSTRACT

AIMS: Ischaemia-reperfusion provokes barrier failure of the coronary microvasculature, impeding functional recovery of the heart during reperfusion. The aim of the present study was to investigate whether the stimulation of cGMP signalling by activation of soluble guanylyl cyclase (sGC) can reduce reperfusion-induced endothelial intercellular gap formation and to determine whether this is due to an influence on endothelial cytosolic Ca(2+) homeostasis during reperfusion. METHODS AND RESULTS: Experiments were performed with cultured coronary endothelial monolayers and isolated saline-perfused rat hearts. HMR1766 (1 micromol/L) or DEAnonoate (0.5 micromol/L) were used to activate sGC. After exposure to simulated ischaemic conditions, reperfusion of endothelial cells led to a pronounced increase in cytosolic calcium levels and intercellular gaps. Stimulation of cGMP signalling during reperfusion increased Ca(2+) sequestration in the endoplasmic reticulum (ER) and attenuated the reperfusion-induced increase in cytosolic [Ca(2+)]. Phosphorylation of phospholamban was also increased, indicating a de-inhibition of the ER Ca(2+) pump (SERCA). Reperfusion-induced intercellular gap formation was reduced. Reduction of myosin light chain phosphorylation indicated inactivation of the endothelial contractile machinery. Effects on cytsolic Ca(2+) and gaps were abrogated by inhibition of cGMP-dependent protein kinase (PKG) with KT5823. In reperfused hearts, stimulation of cGMP signalling led to decreased oedema development. CONCLUSION: sGC/PKG activation during reperfusion reduces reperfusion-induced endothelial intercellular gap formation by attenuation of cytosolic calcium overload and reduction of contractile activation in endothelial cells. This mechanism protects the heart against reperfusion-induced oedema.


Subject(s)
Coronary Vessels/drug effects , Cyclic GMP/metabolism , Endothelial Cells/drug effects , Enzyme Activators/pharmacology , Gap Junctions/drug effects , Myocardial Reperfusion Injury/prevention & control , Receptors, Cytoplasmic and Nuclear/agonists , Second Messenger Systems , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Carbazoles/pharmacology , Cell Hypoxia , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/enzymology , Coronary Vessels/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cytosol/metabolism , Edema, Cardiac/metabolism , Edema, Cardiac/prevention & control , Endoplasmic Reticulum/metabolism , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Gap Junctions/metabolism , Guanylate Cyclase/metabolism , Homeostasis , Hydrazines/pharmacology , Male , Myocardial Reperfusion Injury/metabolism , Myosin Light Chains/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Second Messenger Systems/drug effects , Soluble Guanylyl Cyclase , Sulfonamides/pharmacology , Time Factors , ortho-Aminobenzoates/pharmacology
16.
Eur J Cell Biol ; 88(4): 227-41, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19128857

ABSTRACT

Calcitonin gene-related peptide (CGRP)-alpha is expressed in heart ventricles in sensory nerves and cardiomyocytes. It modifies inotropism and induces ischaemic preconditioning. This study investigates the effect of CGRP-alpha on the contractile responsiveness of isolated adult ventricular rat cardiomyocytes and the effect of chronic hypertension on this interaction. Cardiomyocytes were isolated and paced at 0.5-2.0 Hz. Cell shortening was recorded via a line camera with a reading frame of 500 Hz. CGRP-alpha exerted a dual effect on cardiomyocytes with a positive contractile effect at 10nM and a negative contractile effect at 10 pM. CGRP-alpha(8-37), a calcitonin receptor-like receptor (CRLR) antagonist, attenuated the positive contractile effect. H89, a protein kinase A antagonist, converted the positive contractile effect into a negative contractile effect. The negative contractile effect was converted again back to a positive contractile effect in the presence of l-nitro arginine. In cardiomyocytes isolated from spontaneously hypertensive rats (SHR) the mRNA expression of CRLR and the receptor-associated modifier protein (RAMP)-2 were lower. However, on the protein level CLRL was up-regulated, RAMP2 expression remained unchanged, and eNOS expression was down-regulated in these cells. These cells responded with a pure positive contractile response. In Langendorff preparations, CGRP-alpha slightly reduced the rate pressure product in hearts from normotensive rats but it caused an increase in hearts from SHR. In conclusion, it is shown that CGRP-alpha exerts dual effects on cardiomyocytes favouring the negative contractile effect at very low concentrations. This effect is compensated in chronic pressure-overloaded hearts and converted into a positive inotropism.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , Heart Ventricles/drug effects , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Ventricular Function/drug effects , Animals , Blood Pressure/physiology , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Receptor-Like Protein , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Isoquinolines/pharmacology , Membrane Proteins/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Inbred SHR , Rats, Wistar , Receptor Activity-Modifying Protein 2 , Receptor Activity-Modifying Proteins , Receptors, Calcitonin/antagonists & inhibitors , Sulfonamides/pharmacology , Ventricular Function/physiology
17.
Cardiovasc Res ; 82(1): 77-83, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19168854

ABSTRACT

AIMS: In patients with congestive heart failure, plasma parathyroid hormone (PTH) levels are positively associated with cardiac function. PTH, used to mobilize stem cells from the bone marrow after myocardial infarction, causes an increased left ventricular ejection fraction. The aim of this study was to investigate whether low but plasma-relevant concentrations of PTH directly influence the contractile properties of cardiomyocytes. METHODS AND RESULTS: Isolated adult rat ventricular cardiomyocytes were exposed to PTH(1-34) or full-length PTH at picomolar concentrations for 24 h. Cell shortening was measured at 2 Hz as a cellular correlate of inotropic responsiveness. Intracellular calcium was measured in Fura-AM-loaded cells. PTH(1-3) (20-200 pM) and full-length PTH (200 pM) increased cell shortening within 24 h. PTH had no effect on cell size, but resting and peak systolic calcium concentrations were elevated. The beneficial effect of PTH was mediated via its cAMP/protein kinase A-activating domain and attenuated by addition of a protein kinase A inhibitor. In contrast, PTH peptides representing a protein kinase C-activating domain but not a cAMP/protein kinase A-activating domain or peptides that represent none of these domains had no effect on cell shortening. The effect of PTH on cell shortening was strong at low concentrations of extracellular calcium but declined at higher calcium concentrations. PTH downregulated the expression of the calcium sensing receptor, a receptor known to antagonize the action of PTH on calcium transport. Furthermore, PTH antagonized the angiotensin II-induced loss of cell function. CONCLUSION: Low concentrations of PTH improve cell shortening by increasing calcium load at rest. By this mechanism cardiomyocytes compensate reduced extracellular calcium levels as they occur in patients with heart failure.


Subject(s)
Calcium/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Parathyroid Hormone/metabolism , Angiotensin II/metabolism , Animals , Cardiac Pacing, Artificial , Cell Shape , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Peptide Fragments/metabolism , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Receptors, Calcium-Sensing/metabolism , Signal Transduction , Time Factors
18.
Cardiovasc Res ; 81(1): 159-68, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18806276

ABSTRACT

AIMS: Platelet-derived growth factor BB (PDGF-BB) has been assigned a critical role in vascular growth and recruitment of perivascular mural cells. The purpose of the present study is to investigate the signalling events underlying the stimulation of vasculogenesis of mouse embryonic stem (ES) cells by PDGF-BB. METHODS AND RESULTS: PDGF-BB increased vascular sprouting and branching of capillary-like structures in embryoid bodies as evaluated by computer-assisted analysis of CD31-positive cell structures. It also activated extracellular-regulated kinase 1,2 (ERK1,2) and c-Jun N-terminal kinase but not p38 mitogen-activated protein kinase or PI 3-kinase. Microfluorometric analysis of fluo-4 fluorescence revealed that treatment with PDGF-BB raised intracellular Ca(2+) levels in differentiating ES cells expressing the PDGF receptor beta, an effect that was abolished in the presence of the intracellular Ca(2+) chelator BAPTA. Furthermore, PDGF-BB raised reactive oxygen species (ROS) levels in embryoid bodies as evaluated using the redox-sensitive dye H(2)DCF-DA. ROS generation was blunted in the presence of the NADPH oxidase inhibitors diphenylen iodonium (DPI) and apocynin as well as in the presence of BAPTA, suggesting that ROS generation is regulated by intracellular Ca(2+) transients. The stimulation of vasculogenesis of ES cells upon treatment with PDGF-BB was significantly inhibited by the ERK1,2 inhibitor U0126, the NADPH oxidase inhibitors DPI, apocynin, 4-(2-aminoethyl)benzenesulfonylfluoride and VAS2870, the free radical scavengers vitamin E, and N-(2-mercaptopropionyl)glycin as well as by BAPTA. CONCLUSION: Our data demonstrate that the pro-vasculogenic effects of PDGF-BB are mediated by Ca(2+)-induced ROS generation, resulting in the activation of an ERK1,2-mediated signal transduction cascade.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Calcium/metabolism , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Platelet-Derived Growth Factor/pharmacology , Reactive Oxygen Species/metabolism , Animals , Becaplermin , Calcium Signaling/drug effects , Cell Line , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-sis , Signal Transduction/drug effects
20.
J Cell Physiol ; 217(1): 242-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18528823

ABSTRACT

Pharmacological inhibition of components of the renin-angiotensin-system is one of the major therapeutically options to treat patients with heart failure. This study hypothesized that angiotensin II (Ang II) directly depresses contractile function (cell shortening) by activation of transforming growth factor-beta(1) (TGF-beta(1)). Moreover, we hypothesized that an inhibition of glycogen synthase kinase 3-betaGSK will compensate for this depressive effect by increasing SERCA2 expression. Isolated adult ventricular rat cardiomyocytes were used and cultured in the presence of Ang II (100 nM) for 24 h. Cell shortening and contractile dynamics were recorded at 2 Hz. Immunoblot techniques and gel mobility shift assays were used to demonstrate NFAT activation caused by inhibition of GSK and to demonstrate increases in the expression of SERCA2. Ang-II caused a nearly 20% decrease in cell shortening. This Ang II-dependent effect was mimicked by TGF-beta(1) (10 ng/ml), attenuated by addition of aprotinin, that was used to block the proteolytic activation of TGF-beta(1), or by application of a neutralizing antibody directed against TGF-beta(1). Inhibition of GSK activated NFAT, increased SERCA2 expression and improved cell function. In conclusion, the study identified a paracrine mechanism for the Ang II-dependent loss of cardiac function that occurs independently of hemodynamic changes. Furthermore, it characterized the differences between Ang II and alpha-adrenoceptor stimulation with respect to the maintenance of cellular function explaining cellular events contributing to the difference between adaptive (physiological) and mal-adaptive (patho-physiological) hypertrophy.


Subject(s)
Angiotensin II/pharmacology , Muscle Contraction/drug effects , Myocytes, Cardiac/drug effects , NFATC Transcription Factors/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Male , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/drug effects , Organ Culture Techniques , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...