Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 258
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Gen Intern Med ; 37(14): 3529-3534, 2022 11.
Article in English | MEDLINE | ID: mdl-36042072

ABSTRACT

BACKGROUND: The Veterans Affairs (VA) Healthcare System Rural Transitions Nurse Program (TNP) addresses barriers veterans face when transitioning from urban tertiary VA hospitals to home. Previous clinical evaluations of TNP have shown that enrolled veterans were more likely to follow up with their primary care provider within 14 days of discharge and experience a significant reduction in mortality within 30 days compared to propensity-score matched controls. OBJECTIVE: Examine changes from pre- to post-hospitalization in total, inpatient, and outpatient 30-day healthcare utilization costs for TNP enrollees compared to controls. DESIGN: Quantitative analyses modeling the changes in cost via multivariable linear mixed-effects models to determine the association between TNP enrollment and changes in these costs. PARTICIPANTS: Veterans meeting TNP eligibility criteria who were discharged home following an inpatient hospitalization at one of the 11 implementation sites from April 2017 to September 2019. INTERVENTION: The four-step TNP transitional care intervention. MAIN MEASURES: Changes in 30-day total, inpatient, and outpatient healthcare utilization costs were calculated for TNP enrollees and controls. KEY RESULTS: Among 3001 TNP enrollees and 6002 controls, no statistically significant difference in the change in total costs (p = 0.65, 95% CI: (- $675, $350)) was identified. However, on average, the increase in inpatient costs from pre- to post-hospitalization was approximately $549 less for TNP enrollees (p = 0.02, 95% CI: (- $856, - $246)). The average increase in outpatient costs from pre- to post-hospitalization was approximately $421 more for TNP enrollees compared to controls (p = 0.003, 95% CI: ($109, $671)). CONCLUSIONS: Although we found no difference in change in total costs between veterans enrolled in TNP and controls, TNP was associated with a smaller increase in direct inpatient medical costs and a larger increase in direct outpatient medical costs. This suggests a shifting of costs from the inpatient to outpatient setting.


Subject(s)
Veterans , United States/epidemiology , Humans , United States Department of Veterans Affairs , Patient Acceptance of Health Care , Rural Population , Hospitalization
2.
J Mol Cell Cardiol ; 53(1): 73-81, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22564366

ABSTRACT

AIMS: Since adenine nucleotide translocase 1 (ANT1) overexpression improved cardiac function in rats with activated renin-angiotensin system (RAS) and angiotensin II is known to enhance transforming growth factor ß (TGFß) signaling in cardiomyocytes, we assumed that ANT1 might modulate the classical TGFß/SMAD pathway. We therefore investigated whether the cardioprotective effect of ANT1 overexpression suppresses TGFß(1)-induced apoptosis, whether mitochondrial permeability transition pore (MPTP) regulation is involved, and SMAD signaling pathway is affected. METHODS AND RESULTS: Ventricular cardiomyocytes isolated from wild-type (WT) and ANT1 transgenic rats were treated with the apoptosis-inducing agent TGFß(1) (1 ng/ml). TGFß(1) treatment of WT cells enhanced the number of apoptotic cells by 31.8 ± 11.7% (p<0.01 vs. WT) measured by chromatin condensation. Apoptosis was blocked by 1µM cyclosporine A and by ANT1 overexpression. The protecting effect of ANT1 overexpression on TGFß(1)-induced apoptosis was verified by reduced caspase 3/7 activity and increased Bcl-2 expression. In addition, TGFß(1) decreased mitochondrial membrane potential as measured by JC-1 staining by 18.0 ± 3.7% in WT cardiomyocytes, but only by 7.2 ± 2.8% (p<0.05 vs. WT) in ANT1 cardiomyocytes. Cyclosporine A also attenuated the decline in mitochondrial membrane potential under TGFß(1) in WT cardiomyocytes. Determination of MPTP opening by Calcein assay in isolated cardiomyocytes and calcium retention assay in isolated mitochondria revealed a reduced open probability of MPTP after ANT1 overexpression. In addition to the effects of ANT1 on MPTP opening we investigated if ANT1 may interfere with the classical TGFß signaling pathway. Interestingly, ANT1-transgenic cardiomyocytes expressed less TGFß receptor II than WT cells. However, SMAD2 phosphorylation was already enhanced without TGFß(1) stimulation in these cells. Although no additional increase in SMAD2 phosphorylation was detectable after TGFß(1) treatment, SMAD signaling was still responsive to TGFß(1) indicated by an upregulation of SMAD7, a TGFß(1) target protein. CONCLUSION: Heart-specific overexpression of ANT1 leads to a reduced apoptotic response to TGFß(1) by preservation of the mitochondrial membrane potential, resistance to MPTP opening and altered TGFß signaling.


Subject(s)
Apoptosis/drug effects , Mitochondrial ADP, ATP Translocases/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Apoptosis/genetics , Cells, Cultured , Gene Expression , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Permeability Transition Pore , Rats , Rats, Sprague-Dawley , Signal Transduction , Transgenes
3.
Biochem Biophys Res Commun ; 425(2): 230-6, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22828512

ABSTRACT

BACKGROUND: Apoptotic death of endothelial cells (EC) plays a crucial role for the development of ischemic injury. In the present study we investigated the impact of extracellular Adenosine-5'-triphosphate (ATP), either released from cells or exogenously added, on ischemia-induced apoptosis of human EC. METHODS AND RESULTS: To simulate ischemic conditions, cultured human umbilical vein endothelial cells (HUVEC) were exposed to 2 h of hypoxia (Po(2)<4mm Hg) in serum-free medium. Ischemia led to a 1.7-fold (+/-0.4; P<0.05) increase in EC apoptosis compared to normoxic controls as assessed by immunoblotting and immunocytochemistry of cleaved caspase-3. Ischemia-induced apoptosis was accompanied by a 2.3-fold (+/-0.5; P<0.05) increase of extracellular ATP detected by using a luciferin/luciferase assay. Addition of the soluble ecto-ATPase apyrase, enhancing ATP degradation, increased ischemia-induced caspase-3 cleavage. Correspondingly, inhibition of ATP breakdown by addition of the selective ecto-ATPase inhibitor ARL67156 significantly reduced ischemia-induced apoptosis. Extracellular ATP acts on membrane-bound P2Y- and P2X-receptors to induce intracellular signaling. Both, ATP and the P2Y-receptor agonist UTP significantly reduced ischemia-induced apoptosis in an equipotent manner, whereas the P2X-receptor agonist αß-me-ATP did not alter caspase-3 cleavage. The anti-apoptotic effects of ARL67156 and UTP were abrogated when P2-receptors were blocked by Suramin or PPADS. Furthermore, extracellular ATP led to an activation of MEK/ERK- and PI3K/Akt-signaling pathways. Accordingly, inhibition of MEK/ERK-signaling by UO126 or inhibition of PI3K/Akt-signaling by LY294002 abolished the anti-apoptotic effects of ATP. CONCLUSION: The data of the present study indicate that extracellular ATP counteracts ischemia-induced apoptosis of human EC by activating a P2Y-receptor-mediated signaling reducing caspase-3 cleavage.


Subject(s)
Adenosine Triphosphate/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cytoprotection , Human Umbilical Vein Endothelial Cells/drug effects , Ischemia/enzymology , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/metabolism , Butadienes/pharmacology , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Ischemia/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Morpholines/pharmacology , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism
4.
J Cell Mol Med ; 15(11): 2478-85, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21199327

ABSTRACT

Uncontrolled release of Ca(2+) from the sarcoplasmic reticulum (SR) contributes to the reperfusion-induced cardiomyocyte injury, e.g. hypercontracture and necrosis. To find out the underlying cellular mechanisms of this phenomenon, we investigated whether the opening of mitochondrial permeability transition pores (MPTP), resulting in ATP depletion and reactive oxygen species (ROS) formation, may be involved. For this purpose, isolated cardiac myocytes from adult rats were subjected to simulated ischemia and reperfusion. MPTP opening was detected by calcein release and by monitoring the ΔΨ(m). Fura-2 was used to monitor cytosolic [Ca(2+)](i) or mitochondrial calcium [Ca(2+)](m), after quenching the cytosolic compartment with MnCl(2). Mitochondrial ROS [ROS](m) production was detected with MitoSOX Red and mag-fura-2 was used to monitor Mg(2+) concentration, which reflects changes in cellular ATP. Necrosis was determined by propidium iodide staining. Reperfusion led to a calcein release from mitochondria, ΔΨ(m) collapse and disturbance of ATP recovery. Simultaneously, Ca(2+) oscillations occurred, [Ca(2+)](m) and [ROS](m) increased, cells developed hypercontracture and underwent necrosis. Inhibition of the SR-driven Ca(2+) cycling with thapsigargine or ryanodine prevented mitochondrial dysfunction, ROS formation and MPTP opening. Suppression of the mitochondrial Ca(2+) uptake (Ru360) or MPTP (cyclosporine A) significantly attenuated Ca(2+) cycling, hypercontracture and necrosis. ROS scavengers (2-mercaptopropionyl glycine or N-acetylcysteine) had no effect on these parameters, but reduced [ROS](m). In conclusion, MPTP opening occurs early during reperfusion and is due to the Ca(2+) oscillations originating primarily from the SR and supported by MPTP. The interplay between Ca(2+) cycling and MPTP promotes the reperfusion-induced cardiomyocyte hypercontracture and necrosis. Mitochondrial ROS formation is a result rather than a cause of MPTP opening.


Subject(s)
Calcium/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/physiology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cyclosporine/pharmacology , Fluoresceins/analysis , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondrial Permeability Transition Pore , Necrosis , Rats , Rats, Wistar , Ruthenium Compounds/pharmacology , Ryanodine/pharmacology , Thapsigargin/pharmacology , Tiopronin/pharmacology
5.
J Mol Cell Cardiol ; 48(1): 270-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19406125

ABSTRACT

Ischemic preconditioning has a powerful protective potential against a reperfusion-induced injury of the post-ischemic myocardium. Cardiomyocyte hypercontracture, i.e. excessive cell shortening, is an essential mechanism of the reperfusion-induced injury. Rigor contracture, i.e. Ca(2+)-independent contracture, has been shown to be an import component of the reperfusion-induced hypercontracture. Since rigor contracture is dependent on the rapidity of the metabolic recovery during reoxygenation, we hypothesized that preconditioning of the cardiomyocyte mitochondria may improve mitochondrial function to restore the energy balance during the initial phase of reoxygenation and may thus prevent rigor contracture. For this purpose adult rat cardiomyocytes were exposed to anoxia with subsequent reoxygenation. For preconditioning, cells were pre-treated with the mitochondrial ATP-sensitive K(+) channel opener diazoxide. Pre-treatment with 100 micromol/l diazoxide significantly reduced the reoxygenation-induced hypercontracture of cardiomyocytes due to an attenuation of the Ca(2+)-independent rigor-type contracture, which was accompanied by an acceleration of the phosphocreatine resynthesis during the initial phase of reoxygenation. Treatment with the mitochondrial ATP-sensitive K(+) channel antagonist 5-hydroxydecanoate (500 micromol/l) during preconditioning phase abolished these protective effects. Similarly, partial suppression of the mitochondrial function with 100 micromol/l NaCN during the reoxygenation phase abolished the diazoxide effects. Finally, in isolated rat hearts, preconditioning with diazoxide prior to global ischemia significantly improved left ventricular function and attenuated hypercontracture during reperfusion. This effect could be abolished by the treatment with 100 micromol/l NaCN during reperfusion. Taken together, pharmacological preconditioning of cardiomyocytes with diazoxide protects against the reoxygenation-induced rigor hypercontracture due to an improvement of the energy recovery at the onset of reoxygenation.


Subject(s)
Diazoxide/pharmacology , Ischemic Preconditioning, Myocardial , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology , Hypoxia/physiopathology , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Phosphocreatine/metabolism , Rats , Rats, Wistar , Sodium Cyanide/pharmacology , Vasodilator Agents/pharmacology
6.
Am J Physiol Cell Physiol ; 298(6): C1501-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200209

ABSTRACT

Ischemia-induced apoptosis of endothelial cells may contribute to tissue injury, organ failure, and transplantation rejection. However, little is known about survival mechanisms capable to counteract endothelial apoptosis. This study investigated the potential role of an endogenous anti-apoptotic response elicited by transient hypoxia, capable to avert ongoing apoptosis in endothelial cells. Experiments were carried out in three different types of cultured endothelial cells (human umbilical vein, pig aorta, and from rat coronary microvasculature). As a pro-apoptotic challenge endothelial cells were cultured in serum-free medium and subjected to hypoxia for 2 h. We found that transient hypoxia reduced caspase 3 activation within 1 h of hypoxia. Accordingly, the number of apoptotic cells was reduced after 24 h of reoxygenation. This was true for all three cell types analyzed. Analysis of Akt and mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways revealed that hypoxia induced a transient activation of ERK 2 but not of Akt. ERK 2 phosphorylation preceded the phosphorylation of pro-apoptotic molecule Bad at Ser112, an inhibitory phosphorylation site specific for ERK. The protective effects of hypoxia regarding Bad phosphorylation, caspase 3 activation, and apoptosis were abolished by MEK 1/2 inhibitors, PD98059 or UO126, as well as by antisense oligonucleotides directed against ERK 1/2. Furthermore, inhibition of this pathway inhibited hypoxia-induced increase in mitochondrial membrane potential. The present study demonstrates that transient hypoxia induces a novel survival mechanism that protects endothelial cells against apoptosis. This endogenous process involves MEK/ERK-mediated inhibition of the pro-apoptotic molecule Bad and caspase 3.


Subject(s)
Apoptosis , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Hypoxia , Cell Survival , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , MAP Kinase Kinase Kinases/metabolism , Membrane Potential, Mitochondrial , Oligonucleotides, Antisense/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction , Swine , Time Factors , bcl-Associated Death Protein/metabolism
7.
J Cell Physiol ; 224(1): 120-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20232299

ABSTRACT

Growth differentiation factor 15 (GDF15) is induced during heart failure development, and may influence different processes in cardiac remodeling. While its anti-apoptotic action under conditions of ischemia-reperfusion have been shown, it remained unclear if this is a broadly protective effect applicable to other apoptotic stimuli. Furthermore, effects on cardiac hypertrophy remained obscure. Therefore, we investigated the effects of GDF15 on induction of hypertrophy and apoptosis in ventricular cardiomyocytes. GDF15 (3 ng/ml) enhanced hypertrophic growth of cardiomyocytes as determined by an increase in cell size by 27 +/- 5% and rate of protein synthesis by 47 +/- 15%. In addition, a time and dose-dependent increase in SMAD-binding affinity was found, as well as enhanced phosphorylation of R-SMAD1. Inhibition of SMADs by transformation of cardiomyocytes with SMAD-decoy oligonucleotides abolished the hypertrophic growth effect. Specific inhibitors of PI3K (10 microM LY290042 or 10 nM wortmannin) or ERK (10 microM PD98059) also blocked GDF15-induced hypertrophy and SMAD activation. Apoptosis induction by three different agents, 100 nM angiotensin II, 1 ng/ml TGFbeta(1), or the NO-donor SNAP (100 microM) was blocked by addition of GDF15 (3 ng/ml). Scavenging of SMADs by transformation of cardiomyocytes with SMAD-decoy oligonucleotides abolished the anti-apoptotic effect of GDF15. In conclusion, GDF15 protects ventricular cardiomyocytes against different apoptotic stimuli and enhances hypertrophic growth. Hypertrophic signaling is thereby mediated via the kinases PI3K and ERK and the transcription factor R-SMAD1. Thus, GDF15 may influence cardiac remodeling via two different mechanisms, apoptosis protection and induction of hypertrophy.


Subject(s)
Apoptosis , Cardiomegaly/metabolism , Cardiomegaly/pathology , Growth Differentiation Factor 15/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Signal Transduction , Age Factors , Angiotensin II/metabolism , Animals , Apoptosis/drug effects , Cardiomegaly/prevention & control , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Myocytes, Cardiac/drug effects , Nitric Oxide Donors/pharmacology , Oligonucleotides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Smad1 Protein/genetics , Smad1 Protein/metabolism , Time Factors , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling
8.
J Exp Med ; 189(9): 1373-82, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10224277

ABSTRACT

The effect of factor XIII on endothelial barrier function was studied in a model of cultured monolayers of porcine aortic endothelial cells and saline-perfused rat hearts. The thrombin-activated plasma factor XIII (1 U/ml) reduced albumin permeability of endothelial monolayers within 20 min by 30 +/- 7% (basal value of 5.9 +/- 0.4 x 10(-6) cm/s), whereas the nonactivated plasma factor XIII had no effect. Reduction of permeability to the same extent, i.e., by 34 +/- 9% could be obtained with the thrombin-activated A subunit of factor XIII (1 U/ml), whereas the iodoacetamide-inactivated A subunit as well as the B subunit had no effect on permeability. Endothelial monolayers exposed to the activated factor XIII A exhibited immunoreactive deposition of itself at interfaces of adjacent cells; however, these were not found on exposure to nonactivated factor XIII A or factor XIII B. Hyperpermeability induced by metabolic inhibition (1 mM potassium cyanide plus 1 mM 2-deoxy-D-glucose) was prevented in the presence of the activated factor XIII A. Likewise, the increase in myocardial water content in ischemic-reperfused rat hearts was prevented in its presence. This study shows that activated factor XIII reduces endothelial permeability. It can prevent the loss of endothelial barrier function under conditions of energy depletion. Its effect seems related to a modification of the paracellular passageways in endothelial monolayers.


Subject(s)
Endothelium, Vascular/physiology , Factor XIII/metabolism , Animals , Aorta/cytology , Body Water , Cell Membrane Permeability , Cells, Cultured , Endothelium, Vascular/cytology , Male , Rats , Rats, Wistar , Staining and Labeling , Swine
9.
J Exp Med ; 187(4): 497-503, 1998 Feb 16.
Article in English | MEDLINE | ID: mdl-9463400

ABSTRACT

Anti-neutrophil cytoplasmic antibodies (ANCAs) targeting proteinase 3 (PR3) have a high specifity for Wegener's granulomatosis (WG), and their role in activating leukocytes is well appreciated. In this study, we investigated the influence of PR3-ANCA and murine monoclonal antibodies on human umbilical vascular endothelial cells (HUVECs). Priming of HUVECs with tumor necrosis factor alpha induced endothelial upregulation of PR3 message and surface expression of this antigen, as measured by Cyto-ELISA, with a maximum occurrence after 2 h. Primed cells responded to low concentrations of both antibodies (25 ng-2.5 microg/ml), but not to control immunoglobulins, with pronounced, dose-dependent phosphoinositide hydrolysis, as assessed by accumulation of inositol phosphates. The signaling response peaked after 20 min, in parallel with the appearance of marked prostacyclin and platelet-activating factor synthesis. The F(ab)2 fragment of ANCA was equally potent as ANCA itself. Disrupture of the endothelial F-actin content by botulinum C2 toxin to avoid antigen-antibody internalization did not affect the response. In addition to the metabolic events, anti-PR3 challenge, in the absence of plasma components, provoked delayed, dose-dependent increase in transendothelial protein leakage. We conclude that anti-PR3 antibodies are potent inductors of the preformed phosphoinositide hydrolysis-related signal tranduction pathway in human endothelial cells. Associated metabolic events and the loss of endothelial barrier properties suggest that anti-PR3-induced activation of endothelial cells may contribute to the pathogenetic sequelae of autoimmune vasculitis characterizing WG.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Antibodies, Monoclonal/immunology , Endothelium, Vascular/immunology , Granulomatosis with Polyangiitis/immunology , Serine Endopeptidases/immunology , Signal Transduction , Cell Communication , Cells, Cultured , E-Selectin/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Granulomatosis with Polyangiitis/pathology , Humans , Myeloblastin , Phosphatidylinositols/metabolism , Platelet Activating Factor/metabolism , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism
10.
Cardiovasc Res ; 73(1): 164-71, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17097624

ABSTRACT

OBJECTIVE: Conditions of ischemia-reperfusion disturb the homoeostasis of cytosolic Ca2+ in cardiac microvascular endothelial cells (CMEC), leading to numerous malfunctions of the endothelium. Reperfusion specifically aggravates the Ca2+ overload developed during sustained ischemia. The aim of this study was to identify the origin of the reperfusion-induced part of the Ca2+ overload. Our hypotheses were that this is either due to a Na+-dependent process, e.g. involving the Na+/H+ exchanger (NHE) and/or the Na+/Ca2+ exchanger (NCX), or a process involving the endoplasmic reticulum (ER) and store-operated channels (SOC). METHODS AND RESULTS: Cultured CMEC from rats were exposed to conditions of simulated ischemia (hypoxia, pH 6.4) and reperfusion (reoxygenation, pH 7.4). Cytosolic Ca2+ ([Ca2+]i) and cytosolic Na+ ([Na+]i) concentrations and cytosolic pH (pHi) were measured with the use of fluorescent indicators. Removal of Ca2+ from the extracellular media during reoxygenation prevented the [Ca2+]i rise. Neither the activation of the NHE nor of the NCX in reoxygenated CMEC caused a change in this [Ca2+]i rise. Complete or partial removal of Na+ from the external media also had no effect on the [Ca2+]i rise. In contrast, specific inhibition of the inositol trisphosphate (InsP3) receptor by xestospongin C (3 micromol/l), of phospholipase (PLC) by U73122 (1 micromol/l), or of SOC by the inhibitors gadolinium (10 micromol/l) or 2-APB (50 micromol/l) lowered or abolished the reoxygenation-induced [Ca2+]i rise. CONCLUSION: In CMEC exposed to reperfusion conditions, the enhanced Ca2+ overload is due to Ca2+ influx. The influx is not mediated by a Na+-dependent mechanism, but rather is due to activation of the InsP3 receptor of the ER and activation of SOC.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Ion Channels/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Biological Transport , Cell Hypoxia , Cells, Cultured , Cytosol/metabolism , Endothelial Cells/metabolism , Guanidines/pharmacology , Hydrogen-Ion Concentration , Male , Microcirculation , Ouabain/pharmacology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sodium/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Sulfones/pharmacology , Thapsigargin/pharmacology
11.
Cardiovasc Res ; 74(3): 487-96, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17408601

ABSTRACT

OBJECTIVES: Extracellular ATP stabilizes the endothelial barrier and inactivates the contractile machinery of endothelial cells. This inactivation relies on dephosphorylation of the regulatory myosin light chain (MLC) due to an activation of the MLC phosphatase (MLCP). To date, activation and function of MLCP in endothelial cells are only partially understood. METHODS: Here, the mechanism of extracellular ATP-mediated activation of MLCP was analyzed in human endothelial cells from umbilical veins. Cells were transfected with the endogenous protein phosphatase 1 (PP1)-specific inhibitor-2 (I-2). RESULTS: Overexpression of I-2 led to inhibition of PP1 activity and abrogation of the ATP-induced dephosphorylation of MLC. This indicates that the PP1 catalytic subunit is the principal phosphatase catalyzing the MLC dephosphorylation induced by extracellular ATP. As demonstrated by immunoprecipitation analysis, extracellular ATP recruits the PP1delta catalytic subunit and the myosin phosphatase targeting subunit (MYPT1) to form a complex. ATP stimulated dephosphorylation of MYPT1 at the inhibitory phosphorylation sites threonine 850 and 696. However, extracellular ATP failed to stimulate MYPT1 dephosphorylation in I-2-overexpressing cells. CONCLUSIONS: The present study shows for the first time that, in endothelial cells, extracellular ATP causes activation of MLCP through recruitment of PP1delta and MYPT1 into a MLCP holoenzyme complex and PP1-mediated reduction of the inhibitory phosphorylation of MYPT1.


Subject(s)
Adenosine Triphosphate/pharmacology , Endothelial Cells/enzymology , Myosin-Light-Chain Phosphatase/metabolism , Adenosine Triphosphate/analogs & derivatives , Amides/pharmacology , Blotting, Western , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Marine Toxins , Myosin-Light-Chain Phosphatase/antagonists & inhibitors , Nucleotidases/antagonists & inhibitors , Oxazoles/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Phosphatase 1 , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proteins/genetics , Proteins/metabolism , Purinergic P1 Receptor Antagonists , Pyridines/pharmacology , Theophylline/analogs & derivatives , Theophylline/pharmacology , Thrombin/pharmacology , Transfection/methods , rho-Associated Kinases
12.
Cardiovasc Res ; 73(3): 568-74, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17196568

ABSTRACT

OBJECTIVE: The autonomous proliferative response of endothelial cells to hypoxia has been shown to be dependent on activation of NAD(P)H oxidase, on the cytosolic Ca2+ load, and, consequently, on nuclear translocation of extracellular signal-regulated kinase (ERK)1/2 during transient hypoxia. The aim of the present study was to investigate whether poly(ADP-ribose) polymerase (PARP) is a downstream signal of NAD(P)H oxidase, mediating cytosolic Ca2+ load and hence nuclear translocation of ERK1/2 and endothelial cell proliferation. METHODS: Porcine aortic endothelial cells were incubated under hypoxic conditions for 40 min. Cytosolic [Ca2+] and reactive oxygen species (ROS) formation were measured in fura-2- and DCF-loaded cells, respectively. PARP activation was detected by immunocytochemistry, and endothelial cell proliferation was determined 24 h after 60 min of transient hypoxia. RESULTS: Inhibition of NAD(P)H oxidase with antisense oligonucleotide against the p22(phox) subunit, MEK/ERK signalling with UO 126 (30 microM), or PARP with PJ 34 (10 microM) leads to a marked reduction in hypoxia-induced cytosolic Ca2+ load and activation of PARP. Hypoxia-induced translocation of ERK1/2 and endothelial cell proliferation were also prevented when NAD(P)H oxidase or PARP were inhibited; however, hypoxic ROS formation was not affected in the presence of PARP inhibitor. CONCLUSION: PARP represents a downstream effector of NADP(H) oxidase and acts as a necessary intermediate step for the hypoxic proliferative response of endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular , MAP Kinase Signaling System , Poly(ADP-ribose) Polymerases/physiology , Animals , Butadienes/pharmacology , Calcium/analysis , Calcium/metabolism , Cell Hypoxia/physiology , Cell Proliferation/drug effects , Cells, Cultured , Cytosol/chemistry , Cytosol/metabolism , Endothelial Cells/cytology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Peroxide/metabolism , Immunohistochemistry , Microscopy, Fluorescence , NADPH Oxidases/genetics , Nitriles/pharmacology , Oligonucleotides, Antisense/pharmacology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Reactive Oxygen Species/metabolism , Swine
13.
Neuropsychopharmacology ; 43(12): 2408-2417, 2018 11.
Article in English | MEDLINE | ID: mdl-29773909

ABSTRACT

Amphetamine abuse is a major public health concern for which there is currently no effective treatment. To develop effective treatments, the mechanisms by which amphetamine produces its abuse-related effects need to be fully understood. It is well known that amphetamine exerts its actions by targeting high-affinity transporters for monoamines, in particular the cocaine-sensitive dopamine transporter. Organic cation transporter 3 (OCT3) has recently been found to play an important role in regulating monoamine signaling. However, whether OCT3 contributes to the actions of amphetamine is unclear. We found that OCT3 is expressed in dopamine neurons. Then, applying a combination of in vivo, ex vivo, and in vitro approaches, we revealed that a substantial component of amphetamine's actions is OCT3-dependent and cocaine insensitive. Our findings support OCT3 as a new player in the actions of amphetamine and encourage investigation of this transporter as a potential new target for the treatment of psychostimulant abuse.


Subject(s)
Amphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Octamer Transcription Factor-3/biosynthesis , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
14.
J Mol Med (Berl) ; 84(11): 975-83, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16924465

ABSTRACT

Elevations in angiotensin II (AngII) and transforming growth factor (TGF-beta1) levels are often found under conditions leading to progression of heart failure. From several studies, it is evident that AngII enhances TGF-beta1 expression via activator protein 1 (AP-1) activation, and that this pathway is involved in hypertrophic growth of the heart muscle and in the development of cardiac fibrosis. We now continued characterization of the signaling pathway stimulated by AngII in ventricular cardiomyocytes of rat and analyzed if the enhancement of TGF-beta1 expression by AngII may also contribute to apoptosis induction, which is another predictor of heart failure progression. Stimulation of cardiomyocytes with 100 nM AngII for 2 h activated the transcription factors AP-1 and GATA by 68.6+/-23.9 or 70.7+/-9.8%. Induction of both factors was mediated by p38 mitogen-activated protein kinase (MAPK) because it was totally blocked using a specific inhibitor of the kinase (SB202190). When GATA was inhibited by transformation of cardiomyocytes with decoy oligonucleotides, AngII could not enhance TGF-beta1 expression. This inhibition was observed on the mRNA level in real-time polymerase chain reaction and on the protein level in Western blots. As a consequence, upon AngII stimulation for 24 h, release of TGF-beta1 from cardiomyocytes was also reduced from 240.5+/-50.4 to 130.5+/-22.1% (p<0.05). In contrast to the early induction of GATA and AP-1, the transcription factor similar to mothers against decapentaplegic homolog (SMAD) was induced by AngII after 24 h. This stimulation was dependent on TGF-beta1 because it was blocked by antibodies specific for TGF-beta1. Twenty-four hours after AngII addition, the number of apoptotic cardiomyocytes increased by 6.5+/-1.2%, and this apoptosis induction was blocked when SMAD activity was inhibited by transformation of cardiomyocytes with SMAD decoy oligonucleotides. In conclusion, the transcription factors AP-1 and GATA are activated by p38 MAPK upon AngII stimulation, and both are needed to enhance TGF-beta1 expression in ventricular cardiomyocytes. TGF-beta1 acts in an autocrine loop on the cells to induce apoptosis via SMAD signaling. Thus, the often-found correlation between AngII, TGF-beta1, AP-1, and SMAD in pathogenesis of heart disease reflects the proapoptotic signaling pathway induced by AngII in cardiomyocytes.


Subject(s)
Angiotensin II/metabolism , Apoptosis , Autocrine Communication , MAP Kinase Signaling System , Myocytes, Cardiac/metabolism , Transforming Growth Factor beta1/biosynthesis , Angiotensin II/pharmacology , Animals , Apoptosis/drug effects , Autocrine Communication/drug effects , Blotting, Western , Cardiomyopathies/metabolism , Cells, Cultured , GATA Transcription Factors/biosynthesis , Heart Ventricles/cytology , Heart Ventricles/metabolism , MAP Kinase Signaling System/drug effects , Male , Myocytes, Cardiac/drug effects , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins/metabolism , Time Factors , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Circ Res ; 92(9): 1010-5, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12690038

ABSTRACT

Endothelial cells exhibit an autonomous proliferative response to hypoxia, independent of paracrine effectors. In cultured endothelial cells of porcine aorta, we analyzed the signaling of this response, with a focus on the roles of redox signaling and the MEK/ERK pathway. Transient hypoxia (1 hour) stimulated proliferation by 61+/-4% (n=16; P<0.05 versus control), quantified after 24 hours normoxic postincubation. Hypoxia induced an activation of ERK2 and of NAD(P)H oxidase and a burst of reactive oxygen species (ROS), determined by DCF fluorescence. To inhibit the MEK/ERK pathway, we used PD 98059 (PD, 20 micromol/L); to downregulate NAD(P)H oxidase, we applied p22phox antisense oligonucleotides; and to inhibit mitochondrial ROS generation, we used the ubiquinone derivate mitoQ (MQ, 10 micromol/L). All three inhibitions suppressed the proliferative response: PD inhibited NAD(P)H oxidase activation; p22phox antisense transfection did not inhibit ERK2 activation, but suppressed ROS production; and MQ inhibited ERK2 activation and ROS production. The autonomous proliferative response depends on the MEK/ERK pathway and redox signaling steps upstream and downstream of ERK. Located upstream is ROS generation by mitochondria, downstream is NAD(P)H oxidase.


Subject(s)
Endothelium, Vascular/metabolism , MAP Kinase Signaling System , Membrane Transport Proteins , Reactive Oxygen Species/metabolism , Animals , Cell Division , Cell Hypoxia , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Flavonoids/pharmacology , Free Radical Scavengers/pharmacology , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , NAD/metabolism , NADPH Dehydrogenase/genetics , NADPH Oxidases/physiology , Oligonucleotides, Antisense/genetics , Organophosphorus Compounds/pharmacology , Oxidation-Reduction , Phosphoproteins/genetics , Swine , Ubiquinone/pharmacology
16.
Circ Res ; 86(9): 946-51, 2000 May 12.
Article in English | MEDLINE | ID: mdl-10807866

ABSTRACT

Ventricular cardiomyocytes have previously been identified as potential target cells for parathyroid hormone-related peptide (PTHrP). Synthetic PTHrP peptides exert a positive contractile effect. Because systemic PTHrP levels are normally negligible, this suggests that PTHrP is expressed in the ventricle and acts as a paracrine mediator. We investigated the ventricular expression of PTHrP and its expression in cultured cells isolated from the ventricle, studied the release of PTHrP from hearts and cultures, and investigated whether this authentic PTHrP mimics the biological effects previously described for synthetic PTHrP on ventricular cardiomyocytes. We found PTHrP expressed in ventricles of neonatal and adult rat hearts. In cells isolated from adult hearts, we found PTHrP expression exclusively in coronary endothelial cells but not in cardiomyocytes. The latter, however, are target cells for PTHrP. PTHrP was released from isolated perfused hearts during hypoxic perfusion and from cultured coronary endothelial cells under energy-depleting conditions. This PTHrP was biologically active; ie, it exerted a positive contractile and lusitropic effect on cardiomyocytes. Authentic PTHrP was glycosylated and showed a slightly higher potency than synthetic PTHrP. These results suggest that PTHrP is an endothelium-derived modulator of ventricular function.


Subject(s)
Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Proteins/physiology , Animals , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Endothelium, Vascular/cytology , Heart Ventricles , Hypoxia/metabolism , Male , Myocardial Contraction/physiology , Myocardium/cytology , Myocardium/metabolism , Parathyroid Hormone-Related Protein , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Proteins/chemistry , Proteins/metabolism , Rats , Rats, Wistar , Tissue Distribution
17.
Cardiovasc Res ; 66(1): 123-31, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15769455

ABSTRACT

OBJECTIVE: Reperfusion injury of the myocardium is characterised by development of cardiomyocyte hypercontracture. Previous studies have shown that cGMP-mediated stimuli protect against reperfusion injury, but the cellular mechanism is still unknown. METHODS: To simulate ischemia/reperfusion, adult rat cardiomyocytes were incubated anoxically (pH(o) 6.4) and then reoxygenated (pH(o) 7.4). Cytosolic calcium [Ca(2+)](i) (fura-2 ratio), pH(i) (BCECF ratio), cell length, and phospholamban phosphorylation were analysed. Under simulated ischemia cardiomyocytes develop [Ca(2+)](i) overload. When reoxygenated they rapidly undergo hypercontracture, triggered by oscillations of [Ca(2+)](i). We investigated whether cGMP-mediated stimuli can modulate [Ca(2+)](i) or pH(i) recovery and whether this contributes to their protective effect. Membrane-permeable cGMP analogues, 8-bromo-cGMP (1 mmol/L) or 8-pCPT-cGMP (10 micrommol/L), or a receptor-mediated activator of particulate guanylyl cyclase, urodilatin (1 micromol/L), were applied. RESULTS: The investigated stimuli protect against reoxygenation-induced hypercontracture (cell length as percent of end-ischemic length; control: 68+/-1.6; 8-bromo-cGMP: 88+/-1.5*; 8-pCPT-cGMP: 84+/-2.9*; urodilatin: 87+/-1.1*; n=24; *p<0.05). Recovery from [Ca(2+)](i) overload after 2 min reoxygenation [fura-2 ratio (a.u.); control: 1.43+/-0.15; 8-bromo-cGMP: 1.86+/-0.15*; 8-pCPT-cGMP: 1.92+/-0.19*; urodilatin: 1.93+/-0.24*; n=25; *p<0.05] was accelerated, and the frequency of [Ca(2+)](i) oscillations (min(-1)) was significantly reduced (control: 49+/-5.0 min(-1); 8-bromo-cGMP: 18+/-3.5* min(-1); 8-pCPT-cGMP: 18+/-4.5* min(-1); urodilatin: 16+/-4.1* min(-1); n=24; *p<0.05). cGMP-mediated stimuli increased sarcoplasmic Ca(2+) sequestration (caffeine-releasable Ca(2+) pool: 2-3 fold increase vs. control). Inhibition of sarcoplasmic Ca(2+)-ATPase (SERCA) by thapsigargin (150 nmol/L) or of protein kinase G with KT-5823 (1 micromol/L) abolished the effect of these stimuli on [Ca(2+)](i) recovery. The investigated stimuli significantly enhanced phospholamban phosphorylation. CONCLUSIONS: We conclude that cGMP-dependent signals activate SERCA via a protein kinase G-dependent phosphorylation of phospholamban. The increase in SERCA activity seems to reduce peak [Ca(2+)](i) and [Ca(2+)](i) oscillation during reoxygenation and to attenuate the excessive activation of the contractile machinery that otherwise leads to the development of hypercontracture.


Subject(s)
Calcium/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Carbazoles/pharmacology , Cell Size/drug effects , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cytosol/metabolism , Hydrogen-Ion Concentration , Indoles/pharmacology , Male , Microscopy, Fluorescence , Myocytes, Cardiac/drug effects , Rats , Rats, Wistar , Sarcoplasmic Reticulum/metabolism , Thapsigargin/pharmacology
18.
Biochim Biophys Acta ; 883(3): 531-41, 1986 Oct 01.
Article in English | MEDLINE | ID: mdl-3756219

ABSTRACT

In Ca-tolerant adult cardiomyocytes the contribution of endogenous substrates (glycogen, tri- and diacylglycerol) to oxidative substrate metabolism was investigated. After 4 h in culture medium (M 199 plus 4% fetal calf serum) the cellular triacylglycerol content is 3.6-fold higher than in fresh myocardium and reflects the free fatty acid composition of the medium. When triacylglycerol is degraded, all long-chain fatty acids are hydrolysed at equal rates. In these quiescent cells, the activity of pyruvate dehydrogenase is low (10% of full activity, in Tyrode solution with 5 mM glucose). Up to 30% of full pyruvate dehydrogenase activity, the contribution of non-lipid substrates (glycogen, glucose, lactate and pyruvate) to oxidative energy production is correlated to pyruvate dehydrogenase activity. At 5 mM medium concentration, glucose, lactate and pyruvate share in energy production the proportions of 15, 36 and 50%, whereas endogenous lipolysis accounts for 78, 61 and 46%. It is concluded that these quiescent cardiomyocytes represent cardiac metabolism in a basal state in which the preference for fatty acids, especially from endogenous lipids, is very pronounced. The utilization of endogenous substrates therefore has to be considered in all studies investigating the oxidative metabolism of these isolated cells.


Subject(s)
Lipid Metabolism , Myocardium/metabolism , Animals , Cells, Cultured , Diglycerides/metabolism , Energy Metabolism , Fatty Acids/metabolism , Female , Hydrolysis , Oxidation-Reduction , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Inbred Strains , Triglycerides/metabolism
19.
Biochim Biophys Acta ; 732(1): 193-203, 1983 Jul 13.
Article in English | MEDLINE | ID: mdl-6871190

ABSTRACT

The effects of long-chain fatty acids on mitochondrial functions and red cell stability were studied. In albumin-containing incubation media, fatty acid distribution between the albumin-bound and the unbound fraction was estimated by calculation. When fatty acids are compared to one another on the basis of identical unbound concentrations, their effectiveness differs by orders of magnitude. Fatty acids stimulate mitochondrial basic oxygen consumption, thus lowering the respiratory control index, without changing the ATP/O ratio at lower concentrations. Lower concentrations increase Ca2+ uptake velocity, but decrease maximal Ca2+ storage capacity. The order of effectiveness of different fatty acids is the same for both oxidative phosphorylation and Ca2+ uptake. The influence of fatty acids on red cell stability in hypotonic media is similar to these effects both in concentration range and in order of effectiveness. The influence of fatty acids on red cell stability and their critical micellar concentrations were investigated because these are general characteristics of 'detergent-like' compounds. Critical micellar concentrations of the fatty acids in physiological salt buffers are, in general, at least 10-fold higher than the concentrations exhibiting membrane effects in vitro. Based on these findings it is suggested that, of the various concentrations reported in literature for myocardial non-esterified fatty acids, only the lowest values are physiologically possible.


Subject(s)
Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Fatty Acids, Nonesterified/pharmacology , Intracellular Membranes/metabolism , Mitochondria, Heart/metabolism , Animals , Biological Transport, Active/drug effects , Calcium/metabolism , Erythrocyte Membrane/drug effects , Guinea Pigs , Humans , Intracellular Membranes/drug effects , Micelles , Osmotic Fragility , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Structure-Activity Relationship
20.
J Am Coll Cardiol ; 37(1): 300-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11153756

ABSTRACT

OBJECTIVES: The study investigated whether beta-adrenoceptor antagonists augment the hypertrophic response of cardiomyocytes evoked by norepinephrine. BACKGROUND: In adult ventricular cardiomyocytes, stimulation of alpha- but not beta-adrenoceptors induces myocardial hypertrophy. Natural catecholamines, like norepinephrine, stimulate simultaneously alpha- and beta-adrenoceptors. We investigated whether beta-adrenoceptor stimulation interferes with the hypertrophic response caused by alpha-adrenoceptor stimulation. METHODS: Adult ventricular cardiomyocytes isolated from rats were used as an experimental model. Hypertrophic parameters under investigation were stimulation of phenylalanine incorporation and protein mass, stimulation of 14C-uridine incorporation and RNA mass, and increases in cell shape. RESULTS: Norepinephrine (0.01 to 10 micromol/liter) increased concentration-dependent phenylalanine incorporation; pEC50 value was 5.9 +/- 0.1 (n = 8). The alpha1-adrenoceptor antagonist prazosin (0.1 micromol/liter) suppressed norepinephrine-induced increase in rate of protein synthesis. Conversely, propranolol (1 micromol/liter) and the beta1-adrenoceptor selective antagonists CPG 20712A (300 nmol/liter) or atenolol (1 micromol/liter) augmented increases in phenylalanine incorporation caused by norepinephrine. Addition of the beta2-adrenoceptor antagonist ICI 118,551 (55 nmol/liter) did not influence the hypertrophic effect of norepinephrine. Atenolol augmented the norepinephrine-induced increases of all hypertrophic parameters investigated (i.e., protein mass, uridine incorporation, RNA mass, cell volume, and cross-sectional area). In the presence of norepinephrine, inhibition of beta1-adrenoceptors increased the amount of protein kinase C-alpha and -delta isoforms translocated into the particulate fraction. The effect of pharmacological inhibition of beta1-adrenoceptors could be mimicked by Rp-cAMPS (adenosine-3', 5'-cyclic phosphorothiolate-Rp). The inhibitory effect of beta1-adrenoceptor stimulation on the alpha-adrenoceptor-mediated effect persisted in cardiomyocytes isolated from hypertrophic hearts of rats submitted to aortic banding. CONCLUSIONS: In isolated ventricular cardiomyocytes from rats, beta1-adrenoceptor stimulation attenuates the hypertrophic response evoked by alpha1-adrenoceptor stimulation.


Subject(s)
Cardiomyopathy, Hypertrophic/pathology , Heart Ventricles/drug effects , Norepinephrine/pharmacology , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, beta/drug effects , Animals , Heart Ventricles/pathology , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL