Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Toxicol In Vitro ; 48: 121-127, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29337250

ABSTRACT

Stromal-interaction molecule 1 (STIM1)-mediated store-operated Ca2+ entry (SOCE) plays a key role in mediating cardiomyocyte hypertrophy, both in vitro and in vivo. Moreover, there is growing support for the contribution of SOCE to the Ca2+ overload associated with ischemia/reperfusion injury. Therefore, STIM1 inhibition is proposed as a novel target for controlling both hypertrophy and ischemia/reperfusion-induced Ca2+ overload. Our aim was to evaluate the effect of ML9, a STIM1 inhibitor, on cardiomyocyte viability. ML9 was found to induce cell death in cultured neonatal rat cardiomyocytes. Caspase-3 activation, apoptotic index and release of the necrosis marker lactate dehydrogenase to the extracellular medium were evaluated. ML9-induced cardiomyocyte death was not associated with increased intracellular ROS or decreased ATP levels. Moreover, treatment with ML9 significantly increased levels of the autophagy marker LC3-II, without altering Beclin1 or p62 protein levels. However, treatment with ML9 followed by bafilomycin-A1 did not produce further increases in LC3-II content. Furthermore, treatment with ML9 resulted in decreased LysoTracker® Green staining. Collectively, these data suggest that ML9-induced cardiomyocyte death is triggered by a ML9-dependent disruption of autophagic flux due to lysosomal dysfunction.


Subject(s)
Autophagy/drug effects , Azepines/toxicity , Lysosomes/drug effects , Lysosomes/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Stromal Interaction Molecule 1/antagonists & inhibitors , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Mitochondria/drug effects , Mitochondria/metabolism , Necrosis/chemically induced , Necrosis/pathology , Rats , Reactive Oxygen Species/metabolism
2.
Biochim Biophys Acta Mol Cell Res ; 1865(2): 289-296, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29126879

ABSTRACT

The L-type calcium channel (LTCC) is an important determinant of cardiac contractility. Therefore, changes in LTCC activity or protein levels could be expected to affect cardiac function. Several studies describing LTCC regulation are available, but only a few examine LTCC protein stability. Polycystin-1 (PC1) is a mechanosensor that regulates heart contractility and is involved in mechanical stretch-induced cardiac hypertrophy. PC1 was originally described as an unconventional Gi/o protein-coupled receptor in renal cells. We recently reported that PC1 regulates LTCC stability in cardiomyocytes under stress; however, the mechanism underlying this effect remains unknown. Here, we use cultured neonatal rat ventricular myocytes and hypo-osmotic stress (HS) to model mechanical stretch. The model shows that the Cavß2 subunit is necessary for LTCC stabilization in cardiomyocytes during mechanical stretch, acting through an AKT-dependent mechanism. Our data also shows that AKT activation depends on the G protein-coupled receptor activity of PC1, specifically its G protein-binding domain, and the associated Gßγ subunit of a heterotrimeric Gi/o protein. In fact, over-expression of the human PC1 C-terminal mutant lacking the G protein-binding domain blunted the AKT activation-induced increase in Cav1.2 protein in cardiomyocytes. These findings provide novel evidence that PC1 is involved in the regulation of cardiac LTCCs through a Gißγ-AKT-Cavß2 pathway, suggesting a new mechanism for regulation of cardiac function.


Subject(s)
Calcium Channels, L-Type/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stress, Mechanical , TRPP Cation Channels/metabolism , Animals , Calcium Channels, L-Type/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , TRPP Cation Channels/genetics
3.
Cell Calcium ; 56(5): 390-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25224502

ABSTRACT

Striated muscles (skeletal and cardiac) are major physiological targets of insulin and this hormone triggers complex signaling pathways regulating cell growth and energy metabolism. Insulin increases glucose uptake into muscle cells by stimulating glucose transporter (GLUT4) translocation from intracellular compartments to the cell surface. The canonical insulin-triggered signaling cascade controlling this process is constituted by well-mapped tyrosine, lipid and serine/threonine phosphorylation reactions. In parallel to these signals, recent findings reveal insulin-dependent Ca(2+) mobilization in skeletal muscle cells and cardiomyocytes. Specifically, insulin activates the sarco-endoplasmic reticulum (SER) channels that release Ca(2+) into the cytosol i.e., the Ryanodine Receptor (RyR) and the inositol 1,4,5-triphosphate receptor (IP3R). In skeletal muscle cells, a rapid, insulin-triggered Ca(2+) release occurs through RyR, that is brought about upon S-glutathionylation of cysteine residues in the channel by reactive oxygen species (ROS) produced by the early activation of the NADPH oxidase (NOX2). In cardiomyocytes insulin induces a fast and transient increase in cytoplasmic [Ca(2+)]i trough L-type Ca(2+) channels activation. In both cell types, a relatively slower Ca(2+) release also occurs through IP3R activation, and is required for GLUT4 translocation and glucose uptake. The insulin-dependent Ca(2+) released from IP3R of skeletal muscle also promotes mitochondrial Ca(2+) uptake. We review here these actions of insulin on intracellular Ca(2+) channel activation and their impact on GLUT4 traffic in muscle cells, as well as other implications of insulin-dependent Ca(2+) release from the SER.


Subject(s)
Calcium Signaling/genetics , Diabetes Mellitus/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Diabetes Mellitus/physiopathology , Glucose Transporter Type 4/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle, Skeletal/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
4.
Toxicol In Vitro ; 28(8): 1443-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25149584

ABSTRACT

Cardiac fibroblast (CF) survival is important for the maintenance of the extracellular matrix homeostasis in the heart; providing a functional support to cardiomyocytes necessary for the correct myocardial function. Endoplasmic reticulum (ER) stress causes cellular dysfunction and cell death by apoptosis; and thapsigargin is a well-known ER stress inducer. On the other hand, the chemical chaperone, 4-phenylbutyric acid (4-PBA) had showed to prevent ER stress; however, in cardiac fibroblast both the ER stress induced by thapsigargin and prevention by 4-PBA, have not been studied in detail. Neonate rat CF were treated with thapsigargin in presence or absence of 4-PBA, and cell viability was evaluated by trypan blue exclusion and apoptosis by flow cytometry; whereas CHOP, BIP, PDI, ATF4 and procollagen protein levels were assessed by western blot. In CF, thapsigargin triggered the unfolded protein response detected by early increases in ATF4, CHOP, PDI and BIP protein levels as well as, the accumulation of intracellular procollagen. Thapsigargin also stimulated CF death in a time and concentration-dependent manner. ER stress, CF death and apoptosis induced by thapsigargin were prevented by 4-PBA. Conclusion our data suggest that 4-PBA prevent ER stress, intracellular procollagen accumulation, CF death and apoptosis induced by thapsigargin.


Subject(s)
Phenylbutyrates/pharmacology , Thapsigargin/toxicity , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Fibroblasts/drug effects , Myocytes, Cardiac/cytology , Procollagen/metabolism , Rats , Rats, Sprague-Dawley , Unfolded Protein Response/drug effects
5.
Am J Physiol Endocrinol Metab ; 307(2): E209-24, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24895284

ABSTRACT

Elevating cytosolic Ca(2+) stimulates glucose uptake in skeletal muscle, but how Ca(2+) affects intracellular traffic of GLUT4 is unknown. In tissue, changes in Ca(2+) leading to contraction preclude analysis of the impact of individual, Ca(2+)-derived signals. In L6 muscle cells stably expressing GLUT4myc, the Ca(2+) ionophore ionomycin raised cytosolic Ca(2+) and caused a gain in cell surface GLUT4myc. Extra- and intracellular Ca(2+) chelators (EGTA, BAPTA-AM) reversed this response. Ionomycin activated calcium calmodulin kinase II (CaMKII), AMPK, and PKCs, but not Akt. Silencing CaMKIIδ or AMPKα1/α2 partly reduced the ionomycin-induced gain in surface GLUT4myc, as did peptidic or small molecule inhibitors of CaMKII (CN21) and AMPK (Compound C). Compared with the conventional isoenzyme PKC inhibitor Gö6976, the conventional plus novel PKC inhibitor Gö6983 lowered the ionomycin-induced gain in cell surface GLUT4myc. Ionomycin stimulated GLUT4myc exocytosis and inhibited its endocytosis in live cells. siRNA-mediated knockdown of CaMKIIδ or AMPKα1/α2 partly reversed ionomycin-induced GLUT4myc exocytosis but did not prevent its reduced endocytosis. Compared with Gö6976, Gö6983 markedly reversed the slowing of GLUT4myc endocytosis triggered by ionomycin. In summary, rapid Ca(2+) influx into muscle cells accelerates GLUT4myc exocytosis while slowing GLUT4myc endocytosis. CaMKIIδ and AMPK stimulate GLUT4myc exocytosis, whereas novel PKCs reduce endocytosis. These results identify how Ca(2+)-activated signals selectively regulate GLUT4 exocytosis and endocytosis in muscle cells.


Subject(s)
Calcium Signaling/physiology , Endocytosis , Exocytosis , Glucose Transporter Type 4/metabolism , Muscle Cells/metabolism , Adenylate Kinase/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Endocytosis/drug effects , Exocytosis/drug effects , Ionomycin/pharmacology , Mice , Muscle Cells/drug effects , Protein Kinase C/metabolism , Protein Transport/drug effects
6.
Curr Mol Med ; 13(2): 317-29, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23228132

ABSTRACT

The endoplasmic reticulum has a central role in biosynthesis of a variety of proteins and lipids. Mitochondria generate ATP, synthesize and process numerous metabolites, and are key regulators of cell death. The architectures of endoplasmic reticulum and mitochondria change continually via the process of membrane fusion, fission, elongation, degradation, and renewal. These structural changes correlate with important changes in organellar function. Both organelles are capable of moving along the cytoskeleton, thus changing their cellular distribution. Numerous studies have demonstrated coordination and communication between mitochondria and endoplasmic reticulum. A focal point for these interactions is a zone of close contact between them known as the mitochondrial-associated endoplasmic reticulum membrane (MAM), which serves as a signaling juncture that facilitates calcium and lipid transfer between organelles. Here we review the emerging data on how communication between endoplasmic reticulum and mitochondria can modulate organelle function and determine cellular fate.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Signal Transduction/physiology , Animals , Cell Death , Cell Survival , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Endoplasmic Reticulum/ultrastructure , Humans , Membrane Fusion/physiology , Mitochondria/ultrastructure , Mitochondrial Turnover/physiology , Organelle Size
8.
Cell Death Dis ; 2: e244, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22190003

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide. Although treatments have improved, development of novel therapies for patients with CVD remains a major research goal. Apoptosis, necrosis, and autophagy occur in cardiac myocytes, and both gradual and acute cell death are hallmarks of cardiac pathology, including heart failure, myocardial infarction, and ischemia/reperfusion. Pharmacological and genetic inhibition of autophagy, apoptosis, or necrosis diminishes infarct size and improves cardiac function in these disorders. Here, we review recent progress in the fields of autophagy, apoptosis, and necrosis. In addition, we highlight the involvement of these mechanisms in cardiac pathology and discuss potential translational implications.


Subject(s)
Apoptosis , Myocytes, Cardiac/metabolism , Autophagy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Humans , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Necrosis , Protein Biosynthesis , Stem Cell Transplantation
9.
Mini Rev Med Chem ; 11(5): 390-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21443511

ABSTRACT

Increasing evidence suggests that nongenomic effects of testosterone and anabolic androgenic steroids (AAS) operate concertedly with genomic effects. Classically, these responses have been viewed as separate and independent processes, primarily because nongenomic responses are faster and appear to be mediated by membrane androgen receptors, whereas long-term genomic effects are mediated through cytosolic androgen receptors regulating transcriptional activity. Numerous studies have demonstrated increases in intracellular Ca2+ in response to AAS. These Ca2+ mediated responses have been seen in a diversity of cell types, including osteoblasts, platelets, skeletal muscle cells, cardiac myocytes and neurons. The versatility of Ca2+ as a second messenger provides these responses with a vast number of pathophysiological implications. In cardiac cells, testosterone elicits voltage-dependent Ca2+ oscillations and IP3R-mediated Ca2+ release from internal stores, leading to activation of MAPK and mTOR signaling that promotes cardiac hypertrophy. In neurons, depending upon concentration, testosterone can provoke either physiological Ca2+ oscillations, essential for synaptic plasticity, or sustained, pathological Ca2+ transients that lead to neuronal apoptosis. We propose therefore, that Ca2+ acts as an important point of crosstalk between nongenomic and genomic AAS signaling, representing a central regulator that bridges these previously thought to be divergent responses.


Subject(s)
Anabolic Agents/pharmacology , Androgens/pharmacology , Calcium Signaling/drug effects , Cardiomegaly , Signal Transduction/drug effects , Steroids/pharmacology , Anabolic Agents/adverse effects , Androgens/adverse effects , Cardiomegaly/chemically induced , Humans , Steroids/adverse effects
10.
Endocrinology ; 151(10): 4665-77, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20685879

ABSTRACT

Intracellular calcium levels ([Ca2+]i) and glucose uptake are central to cardiomyocyte physiology, yet connections between them have not been studied. We investigated whether insulin regulates [Ca2+]i in cultured cardiomyocytes, the participating mechanisms, and their influence on glucose uptake via SLC2 family of facilitative glucose transporter 4 (GLUT4). Primary neonatal rat cardiomyocytes were preloaded with the Ca2+ fluorescent dye fluo3-acetoxymethyl ester compound (AM) and visualized by confocal microscopy. Ca2+ transport pathways were selectively targeted by chemical and molecular inhibition. Glucose uptake was assessed using [3H]2-deoxyglucose, and surface GLUT4 levels were quantified in nonpermeabilized cardiomyocytes transfected with GLUT4-myc-enhanced green fluorescent protein. Insulin elicited a fast, two-component, transient increase in [Ca2+]i. Nifedipine and ryanodine prevented only the first component. The second one was reduced by inositol-1,4,5-trisphosphate (IP3)-receptor-selective inhibitors (xestospongin C, 2 amino-ethoxydiphenylborate), by type 2 IP3 receptor knockdown via small interfering RNA or by transfected Gßγ peptidic inhibitor ßARKct. Insulin-stimulated glucose uptake was prevented by bis(2-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid-AM, 2-amino-ethoxydiphenylborate, and ßARK-ct but not by nifedipine or ryanodine. Similarly, insulin-dependent exofacial exposure of GLUT4-myc-enhanced green fluorescent protein was inhibited by bis(2-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid-AM and xestospongin C but not by nifedipine. Phosphatidylinositol 3-kinase and Akt were also required for the second phase of Ca2+ release and GLUT4 translocation. Transfected dominant-negative phosphatidylinositol 3-kinase γ inhibited the latter. In conclusion, in primary neonatal cardiomyocytes, insulin induces an important component of Ca2+ release via IP3 receptor. This component signals to glucose uptake via GLUT4, revealing a so-far unrealized contribution of IP3-sensitive Ca2+ stores to insulin action. This pathway may influence cardiac metabolism in conditions yet to be explored in adult myocardium.


Subject(s)
Glucose Transporter Type 4/metabolism , Glucose/pharmacokinetics , Inositol 1,4,5-Trisphosphate Receptors/physiology , Inositol 1,4,5-Trisphosphate/physiology , Insulin/pharmacology , Myocytes, Cardiac/drug effects , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured , Glucose/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Cardiac/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects
11.
Apoptosis ; 15(8): 887-903, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20454859

ABSTRACT

Hyperosmotic stress promotes rapid and pronounced apoptosis in cultured cardiomyocytes. Here, we investigated if Ca(2+) signals contribute to this response. Exposure of cardiomyocytes to sorbitol [600 mosmol (kg water)(-1)] elicited large and oscillatory intracellular Ca(2+) concentration increases. These Ca(2+) signals were inhibited by nifedipine, Cd(2+), U73122, xestospongin C and ryanodine, suggesting contributions from both Ca(2+) influx through voltage dependent L-type Ca(2+) channels plus Ca(2+) release from intracellular stores mediated by IP(3) receptors and ryanodine receptors. Hyperosmotic stress also increased mitochondrial Ca(2+) levels, promoted mitochondrial depolarization, reduced intracellular ATP content, and activated the transcriptional factor cyclic AMP responsive element binding protein (CREB), determined by increased CREB phosphorylation and electrophoretic mobility shift assays. Incubation with 1 mM EGTA to decrease extracellular [Ca(2+)] prevented cardiomyocyte apoptosis induced by hyperosmotic stress, while overexpression of an adenoviral dominant negative form of CREB abolished the cardioprotection provided by 1 mM EGTA. These results suggest that hyperosmotic stress induced by sorbitol, by increasing Ca(2+) influx and raising intracellular Ca(2+) concentration, activates Ca(2+) release from stores and causes cell death through mitochondrial function collapse. In addition, the present results suggest that the Ca(2+) increase induced by hyperosmotic stress promotes cell survival by recruiting CREB-mediated signaling. Thus, the fate of cardiomyocytes under hyperosmotic stress will depend on the balance between Ca(2+)-induced survival and death pathways.


Subject(s)
Apoptosis/physiology , Calcium Signaling/physiology , Calcium/metabolism , Cell Survival , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Sorbitol/pharmacology , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Indicators and Reagents/pharmacology , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Osmotic Pressure , Rats , Rats, Sprague-Dawley
12.
Cell Death Differ ; 16(7): 1006-17, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19325567

ABSTRACT

The inositol 1,4,5-trisphosphate receptor (IP(3)R) is a major regulator of apoptotic signaling. Through interactions with members of the Bcl-2 family of proteins, it drives calcium (Ca(2+)) transients from the endoplasmic reticulum (ER) to mitochondria, thereby establishing a functional and physical link between these organelles. Importantly, the IP(3)R also regulates autophagy, and in particular, its inhibition/depletion strongly induces macroautophagy. Here, we show that the IP(3)R antagonist xestospongin B induces autophagy by disrupting a molecular complex formed by the IP(3)R and Beclin 1, an interaction that is increased or inhibited by overexpression or knockdown of Bcl-2, respectively. An effect of Beclin 1 on Ca(2+) homeostasis was discarded as siRNA-mediated knockdown of Beclin 1 did not affect cytosolic or luminal ER Ca(2+) levels. Xestospongin B- or starvation-induced autophagy was inhibited by overexpression of the IP(3)R ligand-binding domain, which coimmunoprecipitated with Beclin 1. These results identify IP(3)R as a new regulator of the Beclin 1 complex that may bridge signals converging on the ER and initial phagophore formation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Beclin-1 , Calcium/metabolism , Cell Line , Cell Line, Tumor , Gene Knockdown Techniques , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Membrane Proteins/genetics , Oxazoles/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/metabolism , Rats
13.
Cell Death Differ ; 14(5): 1029-39, 2007 May.
Article in English | MEDLINE | ID: mdl-17256008

ABSTRACT

The reduction of intracellular 1,4,5-inositol trisphosphate (IP(3)) levels stimulates autophagy, whereas the enhancement of IP(3) levels inhibits autophagy induced by nutrient depletion. Here, we show that knockdown of the IP(3) receptor (IP(3)R) with small interfering RNAs and pharmacological IP(3)R blockade is a strong stimulus for the induction of autophagy. The IP(3)R is known to reside in the membranes of the endoplasmic reticulum (ER) as well as within ER-mitochondrial contact sites, and IP(3)R blockade triggered the autophagy of both ER and mitochondria, as exactly observed in starvation-induced autophagy. ER stressors such as tunicamycin and thapsigargin also induced autophagy of ER and, to less extent, of mitochondria. Autophagy triggered by starvation or IP(3)R blockade was inhibited by Bcl-2 and Bcl-X(L) specifically targeted to ER but not Bcl-2 or Bcl-X(L) proteins targeted to mitochondria. In contrast, ER stress-induced autophagy was not inhibited by Bcl-2 and Bcl-X(L). Autophagy promoted by IP(3)R inhibition could not be attributed to a modulation of steady-state Ca(2+) levels in the ER or in the cytosol, yet involved the obligate contribution of Beclin-1, autophagy-related gene (Atg)5, Atg10, Atg12 and hVps34. Altogether, these results strongly suggest that IP(3)R exerts a major role in the physiological control of autophagy.


Subject(s)
Autophagy , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Animals , Autophagy/genetics , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Food Deprivation , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxazoles/pharmacology , Protein Isoforms/metabolism , Rats , bcl-X Protein/metabolism
15.
J Hum Hypertens ; 18(2): 119-25, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14730327

ABSTRACT

Neutral endopeptidase (NEP) hydrolyses angiotensins (Ang) I and II and generates angiotensin-(1-7) [Ang-(1-7)]. In humans, the insertion/deletion (I/D) angiotensin-I converting enzyme (ACE) gene polymorphism determined plasma ACE levels by 40%. In rats, a similar polymorphism determines ACE levels which are inversely associated to NEP activity. The objective of this study is to evaluate the relationship between ACE expression and plasma NEP activity in normotensive subjects and in hypertensive patients. In total, 58 consecutive patients with hypertension, evaluated in our Hypertension Clinic, were compared according to their ACE I/D genotypes with 54 control subjects in terms of both plasma ACE activity and NEP activities. Plasma ACE activity was elevated 51 and 70% in both DD ACE groups (normotensives and hypertensives) compared with their respective ID and II ACE groups (P<0.001). A significant effect of the ACE polymorphism and of the hypertensive status on ACE activity was observed (P<0.001). In normotensive DD ACE subjects, NEP activity was 0.30+/-0.02 U/ml, whereas in the normotensive II ACE and in the normotensive ID ACE subjects NEP activity was increased 65 and 48%, respectively (P<0.001). In the hypertensive DD ACE patients, NEP activity was 0.47+/-0.03 U/mg. An effect of the I/D ACE genotypes on NEP activity (P<0.04) and an interaction effect between the I/D ACE genotype and the hypertensive status were also observed (P<0.001). These results are consistent with a normal and inverse relationship between the ACE polymorphism and NEP activity in normotensive humans (as is also observed in rats). This normal relationship is not observed in hypertensive patients.


Subject(s)
Hypertension/enzymology , Neprilysin/genetics , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Analysis of Variance , Case-Control Studies , DNA/blood , Echocardiography , Female , Genotype , Humans , Hypertension/genetics , Male , Middle Aged , Neprilysin/blood , Peptidyl-Dipeptidase A/blood
16.
Rev Esp Cardiol ; 54(11): 1287-94, 2001 Nov.
Article in Spanish | MEDLINE | ID: mdl-11707239

ABSTRACT

INTRODUCTION: Angiotensin II levels can be partially inhibited during chronic administration of angiotensin converting enzyme (ACE) inhibitors, limiting from a clinical point of view its efficacy in the treatment of hypertension. There are few studies relating ACE activity directly with early prevention of left ventricular hypertrophy (LVH) in systemic hypertension during the administration of an ACE inhibitor (ACEI). AIM: To evaluate the effects of early ACE inhibition with perindopril on the development of hypertension, LVH and levels of angiotensin II (Ang II) in plasma as well as in LV in the rat Goldblatt model (Gb; 2 kidneys-1 clip), 2 weeks after surgery. RESULTS: Systolic blood pressure and relative LV mass increased by 42% and 20% respectively, in the Gb group (p < 0.001). Plasma and LV ACE activities were significantly higher in the Gb rats compared with the control rats. Plasma and LV Ang II levels also increased by 129% and 800%, respectively. Perindorpil prevented hypertension and LVH development by inhibiting plasma ACE (and also LV ACE), and also circulation Ang II in plasma and in the LV. CONCLUSIONS: In this experimental model of hypertensive LVH, there is an early activation of plasma and cardiac ACE. Early administration of an ACE inhibitor prevents the development of hypertension and LVH by inhibiting the increases of plasma and LV Ang II.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Hypertension/complications , Hypertrophy, Left Ventricular/prevention & control , Perindopril/administration & dosage , Angiotensin II/analysis , Angiotensin II/blood , Animals , Antihypertensive Agents/administration & dosage , Drug Evaluation, Preclinical , Hypertension/enzymology , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/etiology , Male , Peptidyl-Dipeptidase A/analysis , Peptidyl-Dipeptidase A/blood , Rats , Rats, Sprague-Dawley
17.
Hypertension ; 38(3 Pt 2): 650-4, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566949

ABSTRACT

Angiotensin I is a substrate for both ACE and for neutral endopeptidase 24.11 (NEP). We hypothesized that high ACE expression is related to low NEP activity. Accordingly, circulating and tissue NEP and ACE activities were measured by fluorometry in homozygous rats (F(0) and F(2)) for the Lewis microsatellite allele (LL, low ACE) and for the Brown Norway microsatellite allele (BB, high ACE). Plasma, lung, and aortic ACE activities in F(0) and F(2) were higher in BB rats than in LL rats (P<0.01), whereas left ventricular ACE activity was similar in both genotypes. In contrast, NEP activity in the LL group was higher in the serum, aorta, and lungs in F(0) and F(2) homozygous (P<0.05). Plasma ACE activity was inversely correlated with serum (r=-0.6 and -0.598 in F(0) and F(2), respectively; P<0.03) and lung NEP activities (r=-0.77 in F(0) and r=-0.59 in F(2), P<0.01). Aortic ACE and NEP activities were also correlated (r=-0.696 and -0.584 in F(0) and F(2), respectively; P<0.03). In conclusion, genetically determined high ACE expression in rats is inversely related to tissue NEP activity, which could determine lower angiotensin-(1-7) tissue levels.


Subject(s)
Endopeptidases/metabolism , Peptidyl-Dipeptidase A/metabolism , Animals , Aorta/enzymology , Blood Pressure/physiology , Endopeptidases/blood , Female , Genotype , Heart Ventricles/enzymology , Lung/enzymology , Male , Peptidyl-Dipeptidase A/blood , Peptidyl-Dipeptidase A/genetics , Polymorphism, Genetic , Rats , Rats, Inbred BN , Rats, Inbred Lew
18.
Cell Tissue Res ; 304(2): 279-85, 2001 May.
Article in English | MEDLINE | ID: mdl-11396721

ABSTRACT

In all cell types, the maintenance of normal cell volume is an essential homeostatic function. Relatively little is known about the induction of apoptosis by hyperosmotic stress and its molecular mechanism in terminally differentiated cardiac myocytes. We compared the apoptotic response of cultured neonatal rat cardiomyoctes to hyperosmotic stress by sorbitol (SOR) with those induced by doxorubicin (Doxo) or angiotensin II (Ang II). We also examined the apoptotic-signaling pathway stimulated by the hyperosmotic stress. Apoptosis was assessed by the observation of: (1) cell viability, (2) DNA fragmentation detected by the TUNEL method and by agarose gel electrophoresis, and (3) poly(ADP-ribose)polymerase (PARP) degradation, and Bcl-XS and Bcl-XL levels by Western blot analysis. Exposure of cardiomyocytes to 0.3 M SOR for 24 h resulted in decreased cell viability and increased generation of oligosomal DNA fragments (2.5-fold of controls). At this time, 83 +/- 5% of SOR-treated myocytes were TUNEL-positive (vs 23.7 +/- 6.8% in controls; P<0.01). PARP levels also decreased by approximately 42% when cardiac myocytes were exposed to SOR. Hyperosmotic stress induced a more rapid and stronger apoptotic response in cardiomyocytes than Doxo or Ang II. In addition, SOR increased 3.2-fold Bcl-XS proapoptotic protein without changes in Bcl-XL antiapoptotic protein levels and in the p53-transactivating activity. Taken together, these results strongly suggest that hyperosmotic stress triggers cardiac myocyte apoptosis in a p53-independent manner, being earlier and stronger than apoptosis induced by Doxo and Ang II.


Subject(s)
Apoptosis/physiology , Heart Ventricles/cytology , Myocardium/cytology , Osmotic Pressure , Proto-Oncogene Proteins c-bcl-2/drug effects , Angiotensin II/pharmacology , Animals , Animals, Newborn , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , DNA Fragmentation/drug effects , Doxorubicin/pharmacology , Kinetics , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Sorbitol/pharmacology , bcl-X Protein
19.
Biochem Biophys Res Commun ; 273(2): 736-44, 2000 Jul 05.
Article in English | MEDLINE | ID: mdl-10873673

ABSTRACT

This study aims to elucidate the signaling pathway for insulin-like growth factor-1 (IGF-1) in cultured neonatal rat cardiomyocytes and particularly the role of IGF-1 in cardiac apoptosis. IGF-1 stimulated polyphosphoinositide turnover, translocation of protein kinase C (PKC) isoforms (alpha, epsilon, and delta) from the soluble to the particulate fraction, activation of phospholipid-dependent and Ca(2+)-, phospholipid-dependent PKC, and activation of the extracellular-regulated kinase (ERK). IGF-1 attenuated sorbitol-induced cardiomyocyte viability and nuclear DNA fragmentation. These antiapoptotic effects of IGF-1 were blocked by PD-098059 (an MEK inhibitor) but not by bisindolylmaleimide I (BIM, a specific PKC inhibitor). The ERK pathway may therefore be an important component in the mechanism whereby IGF-1 exerts its antiapoptotic effect on the cardiomyocyte.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hydrolysis , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocardium/cytology , Myocardium/metabolism , Osmotic Pressure , Phosphatidylinositols/metabolism , Protein Kinase C/antagonists & inhibitors , Rats , Signal Transduction/drug effects
20.
Biochem Biophys Res Commun ; 270(3): 1029-35, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10772945

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is a natural protectant of cardiac myocytes that has been shown to improve cardiac function. The role of IGF-1 in attenuating apoptosis induced by osmotic stress (sorbitol, SOR) or by other known apoptotic stimuli (doxorubicin, angiotensin II, and serum withdrawal) was determined in cultured cardiac myocytes. After 6 h of exposure to SOR, apoptosis was initiated, concomitant with a decrease in cell survival and increases in poly-[ADP-ribose] polymerase (PARP) degradation and DNA fragmentation. These effects were maximal after 24 h. IGF-1 partially attenuated apoptosis induced by sorbitol but not that induced by angiotensin II, doxorubicin, or serum withdrawal. In cells preincubated with IGF-1 before the addition of SOR, we detected an increase in the number of viable cells, a decrease in the generation of DNA fragments on agarose gel electrophoresis and in the percentage of positive TUNEL cells, and a reduction on PARP levels. These results suggest that IGF-1 prevents apoptosis induced by osmotic stress in cardiac myocytes but not apoptosis induced by doxorubicin and angiotensin II.


Subject(s)
Apoptosis/drug effects , Insulin-Like Growth Factor I/pharmacology , Myocardium/cytology , Angiotensin II/pharmacology , Animals , Animals, Newborn , Apoptosis/physiology , Cell Survival/drug effects , Cells, Cultured , Culture Media, Serum-Free , Doxorubicin/pharmacology , Heart/drug effects , Heart Ventricles , Kinetics , Osmolar Concentration , Rats , Rats, Sprague-Dawley , Sorbitol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...