Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
2.
Basic Res Cardiol ; 111(2): 22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26909880

ABSTRACT

The cardiac hormone atrial natriuretic peptide (ANP) regulates systemic and pulmonary arterial blood pressure by activation of its cyclic GMP-producing guanylyl cyclase-A (GC-A) receptor. In the lung, these hypotensive effects were mainly attributed to smooth muscle-mediated vasodilatation. It is unknown whether pulmonary endothelial cells participate in the homeostatic actions of ANP. Therefore, we analyzed GC-A/cGMP signalling in lung endothelial cells and the cause and functional impact of lung endothelial GC-A dysfunction. Western blot and cGMP determinations showed that cultured human and murine pulmonary endothelial cells exhibit prominent GC-A expression and activity which were markedly blunted by hypoxia, a condition known to trigger pulmonary hypertension (PH). To elucidate the consequences of impaired endothelial ANP signalling, we studied mice with genetic endothelial cell-restricted ablation of the GC-A receptor (EC GC-A KO). Notably, EC GC-A KO mice exhibit PH already under resting, normoxic conditions, with enhanced muscularization of small arteries and perivascular infiltration of inflammatory cells. These alterations were aggravated on exposure of mice to chronic hypoxia. Lung endothelial GC-A dysfunction was associated with enhanced expression of angiotensin converting enzyme (ACE) and increased pulmonary levels of Angiotensin II. Angiotensin II/AT1-blockade with losartan reversed pulmonary vascular remodelling and perivascular inflammation of EC GC-A KO mice, and prevented their increment by chronic hypoxia. This experimental study indicates that endothelial effects of ANP are critical to prevent pulmonary vascular remodelling and PH. Chronic endothelial ANP/GC-A dysfunction, e.g. provoked by hypoxia, is associated with activation of the ACE-angiotensin pathway in the lung and PH.


Subject(s)
Atrial Natriuretic Factor/metabolism , Endothelial Cells/metabolism , Hypertension, Pulmonary/enzymology , Hypoxia/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Angiotensin II/metabolism , Animals , Endothelin-1/metabolism , Hypertension, Pulmonary/etiology , Lung/metabolism , Mice , Mice, Knockout , Peptidyl-Dipeptidase A/metabolism , Receptors, Atrial Natriuretic Factor/genetics
3.
Circ Heart Fail ; 7(5): 814-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25027872

ABSTRACT

BACKGROUND: The endocrine balance between atrial natriuretic peptide (ANP) and the renin-angiotensin-aldosterone system is critical for the maintenance of arterial blood pressure and volume homeostasis. This study investigated whether a cardiac imbalance between ANP and aldosterone, toward increased mineralocorticoid receptor (MR) signaling, contributes to adverse left ventricular remodeling in response to pressure overload. METHODS AND RESULTS: We used the MR-selective antagonist eplerenone to test the role of MRs in mediating pressure overload-induced dilatative cardiomyopathy of mice with abolished local, cardiac ANP activity. In response to 21 days of transverse aortic constriction, mice with cardiomyocyte-restricted inactivation (knockout) of the ANP receptor (guanylyl cyclase [GC]-A) or the downstream cGMP-dependent protein kinase I developed enhanced left ventricular hypertrophy and fibrosis together with contractile dysfunction. Treatment with eplerenone (100 mg/kg/d) attenuated left ventricular hypertrophy and fully prevented fibrosis, dilatation, and failure. Transverse aortic constriction induced the cardiac expression of profibrotic connective tissue growth factor and attenuated the expression of SERCA2a (sarcoplasmic reticulum Ca(2+)-ATPase) in knockout mice, but not in controls. These genotype-dependent molecular changes were similarly prevented by eplerenone. ANP attenuated the aldosterone-induced nuclear translocation of MRs via GC-A/cGMP-dependent protein kinase I in transfected HEK 293 (human embryonic kidney) cells. Coimmunoprecipitation and fluorescence resonance energy transfer experiments demonstrated that a population of MRs were membrane associated in close interaction with GC-A and cGMP-dependent protein kinase I and, moreover, that aldosterone caused a conformational change of this membrane MR/GC-A protein complex which was prevented by ANP. CONCLUSIONS: ANP counter-regulates cardiac MR activation in hypertensive heart disease. An imbalance in cardiac ANP/GC-A (inhibition) and aldosterone/MR signaling (augmentation) favors adverse cardiac remodeling in chronic pressure overload.


Subject(s)
Cardiomyopathy, Dilated/genetics , Connective Tissue Growth Factor/genetics , DNA/genetics , Gene Expression Regulation , Myocytes, Cardiac/metabolism , Receptors, Mineralocorticoid/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Animals , Atrial Natriuretic Factor/biosynthesis , Atrial Natriuretic Factor/genetics , Blotting, Western , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Connective Tissue Growth Factor/biosynthesis , Disease Models, Animal , Eplerenone , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Mineralocorticoid Receptor Antagonists/pharmacology , Myocytes, Cardiac/pathology , Receptors, Mineralocorticoid/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/biosynthesis , Signal Transduction/drug effects , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Ventricular Remodeling/drug effects
4.
J Clin Endocrinol Metab ; 98(12): E1988-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24057292

ABSTRACT

BACKGROUND: C-type natriuretic peptide (CNP)/natriuretic peptide receptor 2 (NPR2) signaling is essential for long bone growth. Enhanced CNP production caused by chromosomal translocations results in tall stature, a Marfanoid phenotype, and skeletal abnormalities. A similar phenotype was described in a family with an activating NPR2 mutation within the guanylyl cyclase domain. CASE: Here we describe an extremely tall male without skeletal deformities, with a novel NPR2 mutation (p.Arg655Cys) located in the kinase homology domain. OBJECTIVES: The objective of the study was to investigate the functional and structural effects of the NPR2 mutation. METHODS: Guanylyl cyclase activities of wild-type vs mutant NPR2 were analyzed in transfected human embryonic kidney 293 cells and in skin fibroblasts. The former were also used to study possible interactions between both isoforms. Homology modeling was performed to understand the molecular impact of the mutation. RESULTS: CNP-stimulated cGMP production by the mutant NPR2 was markedly increased in patient skin fibroblasts and transfected human embryonic kidney 293 cells. The stimulatory effects of ATP on CNP-dependent guanylyl cyclase activity were augmented, suggesting that this novel mutation enhances both the responsiveness of NPR2 to CNP and its allosteric modulation/stabilization by ATP. Coimmunoprecipitation showed that wild-type and mutant NPR2 can form stable heterodimers, suggesting a dominant-positive effect. In accordance with augmented endogenous receptor activity, plasma N-terminal pro-CNP (a marker of CNP production in tissues) was reduced in the proband. CONCLUSIONS: We report the first activating mutation within the kinase homology domain of NPR2, resulting in extremely tall stature. Our observations emphasize the important role of this domain in the regulation of guanylyl cyclase activity and bone growth in response to CNP.


Subject(s)
Bone Development , Bone Diseases, Developmental/genetics , Mutation , Receptors, Atrial Natriuretic Factor/genetics , Amino Acid Substitution , Body Height , Bone Diseases, Developmental/metabolism , Bone Diseases, Developmental/pathology , Catalytic Domain , Enzyme Activation , Humans , Male , Middle Aged , Receptors, Atrial Natriuretic Factor/chemistry , Receptors, Atrial Natriuretic Factor/metabolism
5.
Arterioscler Thromb Vasc Biol ; 33(9): 2121-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23814119

ABSTRACT

OBJECTIVE: Histamine increases microvascular endothelial leakage by activation of complex calcium-dependent and -independent signaling pathways. Atrial natriuretic peptide (ANP) via its cGMP-forming guanylyl cyclase-A (GC-A) receptor counteracts this response. Here, we characterized the molecular mechanisms underlying this interaction, especially the role of cGMP-dependent protein kinase I (cGKI). APPROACH AND RESULTS: We combined intravital microscopy studies of the mouse cremaster microcirculation with experiments in cultured microvascular human dermal endothelial cells. In wild-type mice, ANP had no direct effect on the extravasation of fluorescent dextran from postcapillary venules, but strongly reduced the histamine-provoked vascular leakage. This anti-inflammatory effect of ANP was abolished in mice with endothelial-restricted inactivation of GC-A or cGKI. Histamine-induced increases in endothelial [Ca(2+)]i in vitro and of vascular leakage in vivo were markedly attenuated by the Ca(2+)-entry inhibitor SKF96365 and in mice with ablated transient receptor potential canonical (TRPC) 6 channels. Conversely, direct activation of TRPC6 with hyperforin replicated the hyperpermeability responses to histamine. ANP, via cGKI, stimulated the inhibitory phosphorylation of TRPC6 at position Thr69 and prevented the hyperpermeability responses to hyperforin. Moreover, inhibition of cGMP degradation by the phosphodiesterase 5 inhibitor sildenafil prevented the edematic actions of histamine in wild types but not in mice with endothelial GC-A or cGKI deletion. CONCLUSIONS: ANP attenuates the inflammatory actions of histamine via endothelial GC-A/cGMP/cGKI signaling and inhibitory phosphorylation of TRPC6 channels. The therapeutic potential of this novel regulatory pathway is indicated by the observation that sildenafil improves systemic endothelial barrier functions by enhancing the endothelial effects of endogenous ANP.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Calcium/metabolism , Capillary Permeability/drug effects , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Endothelial Cells/drug effects , Histamine Antagonists/pharmacology , Histamine/pharmacology , Microvessels/drug effects , TRPC Cation Channels/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Calcium Channel Blockers/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/deficiency , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Male , Mast Cells/drug effects , Mast Cells/metabolism , Membrane Proteins , Mice , Mice, Knockout , Microvessels/enzymology , Phosphodiesterase 5 Inhibitors/pharmacology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction/drug effects , TRPC Cation Channels/deficiency , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , Time Factors , Transfection
6.
Eur Heart J ; 34(16): 1233-44, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22199120

ABSTRACT

AIMS: Cardiac hypertrophy is a common and often lethal complication of arterial hypertension. Elevation of myocyte cyclic GMP levels by local actions of endogenous atrial natriuretic peptide (ANP) and C-type natriuretic peptide (CNP) or by pharmacological inhibition of phosphodiesterase-5 was shown to counter-regulate pathological hypertrophy. It was suggested that cGMP-dependent protein kinase I (cGKI) mediates this protective effect, although the role in vivo is under debate. Here, we investigated whether cGKI modulates myocyte growth and/or function in the intact organism. METHODS AND RESULTS: To circumvent the systemic phenotype associated with germline ablation of cGKI, we inactivated the murine cGKI gene selectively in cardiomyocytes by Cre/loxP-mediated recombination. Mice with cardiomyocyte-restricted cGKI deletion exhibited unaltered cardiac morphology and function under resting conditions. Also, cardiac hypertrophic and contractile responses to ß-adrenoreceptor stimulation by isoprenaline (at 40 mg/kg/day during 1 week) were unaltered. However, angiotensin II (Ang II, at 1000 ng/kg/min for 2 weeks) or transverse aortic constriction (for 3 weeks) provoked dilated cardiomyopathy with marked deterioration of cardiac function. This was accompanied by diminished expression of the [Ca(2+)]i-regulating proteins SERCA2a and phospholamban (PLB) and a reduction in PLB phosphorylation at Ser16, the specific target site for cGKI, resulting in altered myocyte Ca(2+)i homeostasis. In isolated adult myocytes, CNP, but not ANP, stimulated PLB phosphorylation, Ca(2+)i-handling, and contractility via cGKI. CONCLUSION: These results indicate that the loss of cGKI in cardiac myocytes compromises the hypertrophic program to pathological stimulation, rendering the heart more susceptible to dysfunction. In particular, cGKI mediates stimulatory effects of CNP on myocyte Ca(2+)i handling and contractility.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Cyclic GMP-Dependent Protein Kinase Type I/physiology , Stress, Physiological/physiology , Adrenergic beta-Agonists/pharmacology , Analysis of Variance , Angiotensin II/pharmacology , Animals , Aorta , Blood Pressure/drug effects , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cardiomyopathy, Dilated/genetics , Cardiotonic Agents/pharmacology , Constriction , Cyclic GMP-Dependent Protein Kinase Type I/deficiency , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Echocardiography , Gene Deletion , Hemodynamics/drug effects , Isoproterenol/pharmacology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Natriuretic Peptide, C-Type/physiology , Phosphorylation/physiology , Signal Transduction/physiology , Vasoconstrictor Agents/pharmacology
7.
J Cell Physiol ; 227(2): 686-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21465474

ABSTRACT

Calcium-sensitive potassium channels (K(Ca)3.1) are expressed in virtually all migrating cells. Their activity is required for optimal cell migration so that their blockade leads to slowing down. K(Ca)3.1 channels must be inserted into the plasma membrane in order to elicit their physiological function. However, the plasma membrane of migrating cells is subject to rapid recycling by means of endo- and exocytosis. Here, we focussed on the endocytic internalization and the intracellular transport of the human isoform hK(Ca)3.1. A hK(Ca)3.1 channel construct with an HA-tag in the extracellularly located S3-S4 linker was transfected into migrating transformed renal epithelial MDCK-F cells. Channel internalization was visualized and quantified with immunofluorescence and a cell-based ELISA. Movement of hK(Ca)3.1 channel containing vesicles as well as migration of MDCK-F cells were monitored by means of time lapse video microscopy. hK(Ca)3.1 channels are endocytosed during migration. Most of the hK(Ca)3.1 channel containing vesicles are moving at a speed of up to 2 µm/sec in a microtubule-dependent manner towards the front of MDCK-F cells. Our experiments indicate that endocytosis of hK(Ca)3.1 channels is clathrin-dependent since they colocalize with clathrin adaptor proteins and since it is impaired when a C-terminal dileucine motif is mutated. The C-terminal dileucine motif is also important for the subcellular localization of hK(Ca)3.1 channels in migrating cells. Mutated channels are no longer concentrated at the leading edge. We therefore propose that recycling of hK(Ca)3.1 channels contributes to their characteristic subcellular distribution in migrating cells.


Subject(s)
Cell Movement/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Protein Transport/physiology , Animals , Cell Line , Cell Membrane , Clathrin/metabolism , Dogs , Endocytosis , Gene Expression Regulation/physiology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Mutation , Protein Isoforms
8.
Cardiovasc Res ; 93(1): 141-51, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22025581

ABSTRACT

AIMS: Cardiac atrial natriuretic peptide (ANP) participates in the maintenance of arterial blood pressure and intravascular volume homeostasis. The hypovolaemic effects of ANP result from coordinated actions in the kidney and systemic microcirculation. Hence, ANP, via its guanylyl cyclase-A (GC-A) receptor and intracellular cyclic GMP as second messenger, stimulates endothelial albumin permeability. Ultimately, this leads to a shift of plasma fluid into interstitial pools. Here we studied the role of caveolae-mediated transendothelial albumin transport in the hyperpermeability effects of ANP. METHODS AND RESULTS: Intravital microscopy studies of the mouse cremaster microcirculation showed that ANP stimulates the extravasation of fluorescent albumin from post-capillary venules and causes arteriolar vasodilatation. The hyperpermeability effect was prevented in mice with conditional, endothelial deletion of GC-A (EC GC-A KO) or with deleted caveolin-1 (cav-1), the caveolae scaffold protein. In contrast, the vasodilating effect was preserved. Concomitantly, the acute hypovolaemic action of ANP was abolished in EC GC-A KO and Cav-1(-/-) mice. In cultured microvascular rat fat pad and mouse lung endothelial cells, ANP stimulated uptake and transendothelial transport of fluorescent albumin without altering endothelial electrical resistance. The stimulatory effect on albumin uptake was prevented in GC-A- or cav-1-deficient pulmonary endothelia. Finally, preparation of caveolin-enriched lipid rafts from mouse lung and western blotting showed that GC-A and cGMP-dependent protein kinase I partly co-localize with Cav-1 in caveolae microdomains. CONCLUSION: ANP enhances transendothelial caveolae-mediated albumin transport via its GC-A receptor. This ANP-mediated cross-talk between the heart and the microcirculation is critically involved in the regulation of intravascular volume.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Atrial Natriuretic Factor/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Albumins/metabolism , Animals , Caveolae/drug effects , Caveolae/physiology , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/physiology , Cells, Cultured , Endocytosis/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/physiology , Transcytosis/drug effects
9.
Proc Natl Acad Sci U S A ; 108(45): 18500-5, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-22027011

ABSTRACT

Cardiac atrial natriuretic peptide (ANP) regulates arterial blood pressure, moderates cardiomyocyte growth, and stimulates angiogenesis and metabolism. ANP binds to the transmembrane guanylyl cyclase (GC) receptor, GC-A, to exert its diverse functions. This process involves a cGMP-dependent signaling pathway preventing pathological [Ca(2+)](i) increases in myocytes. In chronic cardiac hypertrophy, however, ANP levels are markedly increased and GC-A/cGMP responses to ANP are blunted due to receptor desensitization. Here we show that, in this situation, ANP binding to GC-A stimulates a unique cGMP-independent signaling pathway in cardiac myocytes, resulting in pathologically elevated intracellular Ca(2+) levels. This pathway involves the activation of Ca(2+)-permeable transient receptor potential canonical 3/6 (TRPC3/C6) cation channels by GC-A, which forms a stable complex with TRPC3/C6 channels. Our results indicate that the resulting cation influx activates voltage-dependent L-type Ca(2+) channels and ultimately increases myocyte Ca(2)(+)(i) levels. These observations reveal a dual role of the ANP/GC-A-signaling pathway in the regulation of cardiac myocyte Ca(2+)(i) homeostasis. Under physiological conditions, activation of a cGMP-dependent pathway moderates the Ca(2+)(i)-enhancing action of hypertrophic factors such as angiotensin II. By contrast, a cGMP-independent pathway predominates under pathophysiological conditions when GC-A is desensitized by high ANP levels. The concomitant rise in [Ca(2+)](i) might increase the propensity to cardiac hypertrophy and arrhythmias.


Subject(s)
Atrial Natriuretic Factor/metabolism , Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Myocardium/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Signal Transduction , Animals , Cell Line , Fluorescence Resonance Energy Transfer , Humans , Mice
10.
Endocrinology ; 151(8): 3665-74, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20555029

ABSTRACT

Atrial natriuretic peptide (ANP) and its guanylyl cyclase-A (GC-A) receptor are being involved in metabolism, although their role in the endocrine pancreas is still greatly unknown. The aim of this work is to study a possible role for the ANP/GC-A system in modulating pancreatic beta-cell function. The results presented here show a direct effect of the GC-A receptor in regulating glucose-stimulated insulin secretion (GSIS) and beta-cell mass. GC-A activation by its natural ligand, ANP, rapidly blocked ATP-dependent potassium (K(ATP)) channel activity, increased glucose-elicited Ca(2+) signals, and enhanced GSIS in islets of Langerhans. The effect in GSIS was inhibited in islets from GC-A knockout (KO) mice. Pancreatic islets from GC-A KO mice responded to increasing glucose concentrations with enhanced insulin secretion compared with wild type (WT). Remarkably, islets from GC-A KO mice were smaller, presented lower beta-cell mass and decreased insulin content. However, glucose-induced Ca(2+) response was more vigorous in GC-A KO islets, and basal K(ATP) channel activity in GC-A KO beta-cells was greatly diminished compared with WT. When protein levels of the two K(ATP) channel constitutive subunits sulfonylurea receptor 1 and Inward rectifier potassium channel 6.2 were measured, both were diminished in GC-A KO islets. These alterations on beta-cell function were not associated with disruption of glucose tolerance or insulin sensitivity in vivo. Glucose and insulin tolerance tests were similar in WT and GC-A KO mice. Our data suggest that the ANP/GC-A system may have a modulating effect on beta-cell function.


Subject(s)
Atrial Natriuretic Factor/physiology , Insulin-Secreting Cells/physiology , Receptors, Atrial Natriuretic Factor/physiology , Animals , Atrial Natriuretic Factor/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Female , Glucose/metabolism , Glucose/pharmacology , Insulin/analysis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , KATP Channels/drug effects , KATP Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism
11.
FEBS J ; 277(11): 2440-53, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20456499

ABSTRACT

Atrial natriuretic peptide (ANP), via its guanylyl cyclase A (GC-A) receptor and intracellular guanosine 3',5'-cyclic monophosphate production, is critically involved in the regulation of blood pressure. In patients with chronic heart failure, the plasma levels of ANP are increased, but the cardiovascular actions are severely blunted, indicating a receptor or postreceptor defect. Studies on metabolically labelled GC-A-overexpressing cells have indicated that GC-A is extensively phosphorylated, and that ANP-induced homologous desensitization of GC-A correlates with receptor dephosphorylation, a mechanism which might contribute to a loss of function in vivo. In this study, tandem MS analysis of the GC-A receptor, expressed in the human embryonic kidney cell line HEK293, revealed unambiguously that the intracellular domain of the receptor is phosphorylated at multiple residues: Ser487, Ser497, Thr500, Ser502, Ser506, Ser510 and Thr513. MS quantification based on multiple reaction monitoring demonstrated that ANP-provoked desensitization was accompanied by a complex pattern of receptor phosphorylation and dephosphorylation. The population of completely phosphorylated GC-A was diminished. However, intriguingly, the phosphorylation of GC-A at Ser487 was selectively enhanced after exposure to ANP. The functional relevance of this observation was analysed by site-directed mutagenesis. The substitution of Ser487 by glutamate (which mimics phosphorylation) blunted the activation of the GC-A receptor by ANP, but prevented further desensitization. Our data corroborate previous studies suggesting that the responsiveness of GC-A to ANP is regulated by phosphorylation. However, in addition to the dephosphorylation of the previously postulated sites (Ser497, Thr500, Ser502, Ser506, Ser510), homologous desensitization seems to involve the phosphorylation of GC-A at Ser487, a newly identified site of phosphorylation. The identification and further characterization of the specific mechanisms involved in the downregulation of GC-A responsiveness to ANP may have important pathophysiological implications.


Subject(s)
Cyclic GMP/metabolism , Guanylate Cyclase/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Amino Acid Sequence , Animals , Atrial Natriuretic Factor/blood , Cardiomegaly/blood , Catalytic Domain , Cell Line , Heart Failure/blood , Humans , Kidney/embryology , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/pharmacology , Oligopeptides , Peptides/physiology , Phosphopeptides/analysis , Phosphorylation , Rats , Second Messenger Systems/physiology
12.
J Physiol ; 588(Pt 2): 325-39, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19948658

ABSTRACT

Atrial natriuretic peptide (ANP) via its guanylyl cyclase-A (GC-A) receptor participates in regulation of arterial blood pressure and vascular volume. Previous studies demonstrated that concerted renal diuretic/natriuretic and endothelial permeability effects of ANP cooperate in intravascular volume regulation. We show that the microvascular endothelial contribution to the hypovolaemic action of ANP can be measured by the magnitude of the ANP-induced increase in blood-to-tissue albumin transport, measured as plasma albumin clearance corrected for intravascular volume change, relative to the corresponding increase in ANP-induced renal water excretion. We used a two-tracer method with isotopically labelled albumin to measure clearances in skin and skeletal muscle of: (i) C57BL6 mice; (ii) mice with endothelium-restricted deletion of GC-A (floxed GC-A x tie2-Cre: endothelial cell (EC) GC-A knockout (KO)); and (iii) control littermates (floxed GC-A mice with normal GC-A expression levels). Comparison of albumin clearances in hypervolaemic EC GC-A KO mice with normovolaemic littermates demonstrated that skeletal muscle albumin clearance with ANP treatment accounts for at most 30% of whole body clearance required for ANP to regulate plasma volume. Skin microcirculation responded to ANP similarly. Measurements of permeability to a high molecular mass contrast agent (35 kD Gadomer) by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) enabled repeated measures in individual animals and confirmed small increases in muscle and skin microvascular permeability after ANP. These quantitative methods will enable further evaluation of the contribution of ANP-dependent microvascular beds (such as gastro-intestinal tract) to plasma volume regulation.


Subject(s)
Albumins/metabolism , Atrial Natriuretic Factor/pharmacology , Capillary Permeability/physiology , Muscle, Skeletal/metabolism , Plasma Volume/physiology , Receptors, Atrial Natriuretic Factor/physiology , Skin/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Capillary Permeability/drug effects , Female , Magnetic Resonance Imaging , Mice , Mice, Knockout , Microcirculation/drug effects , Microcirculation/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Plasma Volume/drug effects , Skin/blood supply , Skin/drug effects , Time Factors
13.
Mol Endocrinol ; 23(12): 1973-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19855088

ABSTRACT

The ATP-sensitive potassium (K(ATP)) channel is a key molecule involved in glucose-stimulated insulin secretion. The activity of this channel regulates beta-cell membrane potential, glucose- induced [Ca(2+)](i) signals, and insulin release. In this study, the rapid effect of physiological concentrations of 17beta-estradiol (E2) on K(ATP) channel activity was studied in intact beta-cells by use of the patch-clamp technique. When cells from wild-type (WT) mice were used, 1 nm E2 rapidly reduced K(ATP) channel activity by 60%. The action of E2 on K(ATP) channel was not modified in beta-cells from ERalpha-/- mice, yet it was significantly reduced in cells from ERbeta-/- mice. The effect of E2 was mimicked by the ERbeta agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN). Activation of ERbeta by DPN enhanced glucose-induced Ca(2+) signals and insulin release. Previous evidence indicated that the acute inhibitory effects of E2 on K(ATP) channel activity involve cyclic GMP and cyclic GMP-dependent protein kinase. In this study, we used beta-cells from mice with genetic ablation of the membrane guanylate cyclase A receptor for atrial natriuretic peptide (also called the atrial natriuretic peptide receptor) (GC-A KO mice) to demonstrate the involvement of this membrane receptor in the rapid E2 actions triggered in beta-cells. E2 rapidly inhibited K(ATP) channel activity and enhanced insulin release in islets from WT mice but not in islets from GC-A KO mice. In addition, DPN reduced K(ATP) channel activity in beta-cells from WT mice, but not in beta-cells from GC-A KO mice. This work unveils a new role for ERbeta as an insulinotropic molecule that may have important physiological and pharmacological implications.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor beta/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , KATP Channels/drug effects , KATP Channels/metabolism , Receptors, Atrial Natriuretic Factor/physiology , Animals , Calcium/metabolism , Cells, Cultured , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Female , Immunohistochemistry , Mice , Mice, Knockout , Nitriles/pharmacology , Patch-Clamp Techniques , Phenols , Pyrazoles/pharmacology , Receptors, Atrial Natriuretic Factor/genetics
14.
J Biol Chem ; 283(42): 28313-20, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18713751

ABSTRACT

Atrial (ANP) and B-type natriuretic peptides (BNP) modulate blood pressure and volume through the stimulation of cyclic GMP production by their guanylyl cyclase-A (GC-A) receptor. A novel isoform of GC-A has been identified that is the result of differential splicing of exon 4. The deletion of a 51-bp sequence is predicted to delete 17 amino acids (Lys314-Gln330) in the membrane-distal part of the extracellular domain. Reverse transcription-PCR analyses demonstrated low messenger RNA expression levels of spliced GC-A in all tissues. Homology modeling suggested that the alterations in the protein structure could interfere with ANP binding or signaling. Indeed, functional studies in transfected HEK 293 cells demonstrated that binding of ANP and ANP-induced cyclic GMP formation by GC-ADelta(Lys314-Gln330) were totally abolished. Furthermore, cotransfection studies showed that this GC-A variant forms heterodimers with the wild type receptor and inhibits ligand-inducible cGMP generation. Finally, treatment of mice with angiotensin II (300 ng/kg/min during 7 days) resulted in enhanced pulmonary mRNA expression of spliced GC-A, which was concomitant to diminished GC-A/cGMP responses to ANP. We conclude that alternative splicing can regulate endogenous ANP/GC-A signaling. Angiotensin II-induced alternative splicing of GC-A may represent a novel mechanism for reducing the sensitivity to ANP.


Subject(s)
Alternative Splicing , Atrial Natriuretic Factor/chemistry , Atrial Natriuretic Factor/physiology , Receptors, Atrial Natriuretic Factor/chemistry , Receptors, Atrial Natriuretic Factor/physiology , Angiotensin II/metabolism , Animals , Atrial Natriuretic Factor/genetics , Cell Line , Cyclic GMP/metabolism , Humans , Lung/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Peptides/chemistry , Rats , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction
15.
Endocrinology ; 149(8): 4193-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450968

ABSTRACT

Atrial natriuretic peptide (ANP) regulates arterial blood pressure and volume. Its guanylyl cyclase-A (GC-A) receptor is expressed in vascular endothelium and mediates increases in cGMP, but the functional relevance is controversial. Notably, mice with endothelial-restricted GC-A deletion [EC GC-A knockout (KO) mice] exhibit significant chronic hypervolemic hypertension. The present study aimed to characterize the endothelial effects of ANP and their relevance for the acute regulation of intravascular fluid volume. We studied the effect of ANP on microvascular permeability to fluorescein isothiocyanate-labeled albumin (BSA) using intravital microscopy on mouse dorsal skinfold chambers. Local superfusion of ANP (100 nm) increased microvascular fluorescein isothiocyanate-BSA extravasation in control but not EC GC-A KO mice. Intravenous infusion of synthetic ANP (500 ng/kg x min) caused immediate increases in hematocrit in control mice, indicating intravascular volume contraction. In EC GC-A KO mice, the hematocrit responses were not only abolished but even reversed. Furthermore, acute vascular volume expansion, which caused release of endogenous cardiac ANP, did not affect resting central venous pressure of control mice but rapidly and significantly increased central venous pressure of EC GC-A KO mice. In cultured lung endothelial cells, ANP provoked cGMP-dependent protein kinase I-mediated phosphorylation of vasodilator-stimulated phosphoprotein. We conclude that ANP, via GC-A, enhances microvascular endothelial macromolecule permeability in vivo. This effect might be mediated by cGMP-dependent protein kinase I-dependent phosphorylation of vasodilator-stimulated phosphoprotein. Modulation of transcapillary protein and fluid transport may represent one of the most important hypovolemic actions of ANP.


Subject(s)
Blood Volume/physiology , Cell Communication/physiology , Endothelium, Vascular/physiology , Heart/physiology , Receptors, Atrial Natriuretic Factor/physiology , Animals , Atrial Natriuretic Factor/blood , Atrial Natriuretic Factor/pharmacology , Blood Volume/genetics , Capillary Permeability/drug effects , Capillary Permeability/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hematocrit , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Myocardium/metabolism , Phosphoproteins/metabolism , Phosphorylation , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Venous Pressure/genetics
16.
Adv Exp Med Biol ; 614: 157-64, 2008.
Article in English | MEDLINE | ID: mdl-18290326

ABSTRACT

Tumor cells exhibit mechanisms by which chemotherapeutic drugs can be actively pumped out of the cell (e.g., p-glycoprotein pGP, MRP1), resulting in a multidrug resistant phenotype. Many human tumors show pronounced hypoxia which can result in a local ATP depletion which in turn may compromise the efficacy of these transporters. The aim of this study was therefore to assess the transport activity and expression of drug transporters under hypoxic conditions. Prostate carcinoma cells (R3327-AT1) were exposed to hypoxia (pO2 < 0.5 mmHg) for up to 24h and pump activity was determined by an efflux assay. The results showed that exposing cells to hypoxia for 3-6 h led to a moderate increase in pGP activity. After 24 h pGP activity was reduced by 44% compared to control levels. Hypoxia reduced the MRPI activity to a lesser extent (by 25%). However, the expression of pGP and MRP1 was almost independent of the medium pO2. In conclusion, pronounced hypoxia had only minor effects on the activity of drug transporters with the activity decreasing only after 12-24 h under hypoxia, possibly as a result of ATP depletion. Instead, indirect effects of hypoxia leading to extracellular acidosis seem to have a much more pronounced effect on pGP activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carcinoma/metabolism , Cell Hypoxia/physiology , Multidrug Resistance-Associated Proteins/metabolism , Prostatic Neoplasms/metabolism , Acidosis/metabolism , Animals , Biological Transport , Carcinoma/pathology , Cell Line, Tumor , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Isotonic Solutions , Male , Prostatic Neoplasms/pathology , Rats , Rhodamine 123/metabolism , Ringer's Solution , Time Factors , Verapamil/pharmacokinetics
17.
Adv Exp Med Biol ; 599: 155-61, 2007.
Article in English | MEDLINE | ID: mdl-17727260

ABSTRACT

In the microenvironment of solid growing tumors, pronounced hypoxia or extracellular acidosis is commonly found. The aim of this study was the analysis of the cytotoxic effect of different chemotherapeutic agents (cisplatin, daunorubicin, docetaxel) under these conditions in vitro. Prostate carcinoma cells (R3327-AT1) were exposed to hypoxia (pO2 < 0.5 mmHg) or extracellular acidosis (pH = 6.6) for 6h. After 3h, cytotoxic drugs were added. The cytotoxic effect was assessed by measuring caspase 3-activity (apoptosis), LDH release (necrosis) and repopulation of the cells after chemotherapy (cell death). Compared to aerobic control conditions, severe hypoxia over 6 h per se led to a slight increase in apoptosis, necrosis and cell death. With all three chemotherapeutic agents, hypoxia led to a reduced (by approx. 25%) caspase 3-activity and a marked increase in necrosis. However, the overall cytotoxicity of the drug was not affected by O2-deficiency. By contrast, during extracellular acidosis, the cytotoxic effect of daunorubicin was reduced by 40%, preferentially due to a marked reduction in apoptosis. With cisplatin and docetaxel no change in overall cell death was detected. However, for daunorubicin the tumor-pH seems to have a strong impact on cytotoxicity. With this chemotherapeutic drug the therapeutic efficacy is markedly reduced in an acidotic environment.


Subject(s)
Acidosis , Antineoplastic Agents/therapeutic use , Extracellular Space/chemistry , Hypoxia , Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cisplatin/metabolism , Cisplatin/therapeutic use , Daunorubicin/metabolism , Daunorubicin/therapeutic use , Docetaxel , Humans , Hydrogen-Ion Concentration , Neoplasms/pathology , Rats , Taxoids/metabolism , Taxoids/therapeutic use
18.
Endocrinology ; 148(9): 4162-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17510245

ABSTRACT

The cardiac hormones atrial and brain natriuretic peptides (NPs) counteract the systemic, hypertensive, and hypervolemic actions of angiotensin II (Ang II) via their guanylyl cyclase-A (GC-A) receptor. In the present study, we took advantage of genetically modified mice with conditional, cardiomyocyte (CM)-restricted disruption of GC-A (CM GC-A knockout mice) to study whether NPs can moderate not only the endocrine but also the cardiac actions of Ang II in vivo. Fluorometric measurements of [Ca(2+)](i) transients in isolated, electrically paced adult CMs showed that atrial NP inhibits the stimulatory effects of Ang II on free cytosolic Ca(2+) transients via GC-A. Remarkably, GC-A-deficient CMs exhibited greatly enhanced [Ca(2+)](i) responses to Ang II, which was partly related to increased activation of the Na(+)/H(+)-exchanger NHE-1. Chronic administration of Ang II to control and CM GC-A knockout mice (300 ng/kg body weight per minute via osmotic minipumps during 2 wk) provoked significant cardiac hypertrophy, which was markedly exacerbated in the later genotype. This was concomitant to increased cardiac expression of NHE-1 and enhanced activation of the Ca(2+)/calmodulin-dependent prohypertrophic signal transducers Ca(2+)/calmodulin-dependent kinase II and calcineurin. On the basis of these results, we conclude that NPs exert direct local, GC-A-mediated myocardial effects to antagonize the [Ca(2+)](i)-dependent hypertrophic growth response to Ang II.


Subject(s)
Angiotensin II/pharmacology , Atrial Natriuretic Factor/physiology , Heart/physiology , Receptors, Atrial Natriuretic Factor/deficiency , Ventricular Remodeling/physiology , Angiotensin II/antagonists & inhibitors , Animals , Atrial Natriuretic Factor/genetics , Blood Pressure/drug effects , Calcium Signaling/physiology , Heart Ventricles/drug effects , Mice , Mice, Knockout , Organ Size , RNA, Messenger/genetics , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/physiology , Ventricular Remodeling/drug effects
19.
Oncol Rep ; 17(1): 239-44, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143504

ABSTRACT

The metabolic microenvironment of solid tumors is characterized by an oxygen deficiency and increased anaerobic glycolysis leading to extracellular acidosis and ATP depletion, which in turn may affect other energy-dependent cellular pathways. Since many tumors overexpress active drug transporters (e.g. the p-glycoprotein) leading to a multidrug-resistant phenotype, this study analyzes the impact of the different aspects of the extracellular microenvironment (hypoxia and acidosis) on the activity and expression of the p-glycoprotein (pGP) in the human colon carcinoma cell line LS513. For up to 24 h cells were exposed to hypoxia (pO2<0.5 mmHg), an acidic extracellular environment (pH 6.6), or the combination of hypoxia and acidosis. Under hypoxic conditions (at a normal pH), the pGP activity (measured by the daunorubicin efflux) and the pGP expression were not markedly altered. Under acidic conditions, however, the pGP-mediated drug efflux was increased, an effect which was even more pronounced when the cells were exposed to hypoxia and acidosis simultaneously (increasing the pGP-activity by 70%). The cellular pGP expression remained almost constant under these conditions, indicating that the increased transport rate results from a functional modulation. The findings of the present study indicate that the parameters of the tumor microenviroment (especially extracellular acidosis) can increase the pGP-mediated drug efflux, an effect which may explain the reduced cytotoxicity of chemotherapeutic agents in hypoxic/acidic tumors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Colorectal Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Acidosis/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Daunorubicin/pharmacokinetics , Humans , Hydrogen-Ion Concentration
20.
Neoplasia ; 8(2): 143-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16611407

ABSTRACT

The expression and activity of P-glycoprotein (pGP) play a role in the multidrug resistance of tumors. Because solid-growing tumors often show pronounced hypoxia or extracellular acidosis, this study attempted to analyze the impact of an acidic environment on the expression and activity of pGP and on the cytotoxicity of chemotherapeutic agents. For this, prostate carcinoma cells were exposed to an acidic extracellular environment (pH 6.6) for up to 24 hours. pGP activity was more than doubled after 3 to 6 hours of incubation in acidic medium, whereas cellular pGP expression remained constant, indicating that increased transport rate is the result of functional modulation. In parallel, the cytotoxic efficacy of daunorubicin showed pronounced reduction at low pH, an effect that was reversible on coincubation with a pGP inhibitor. A reduction of intracellular Ca2+ concentration by 35% under acidic conditions induced a higher transport rate of pGP, an effect comparable to that found on inhibition of protein kinase C (PKC). These data indicate that pGP activity is increased by low extracellular pH presumably as a result of lowered intracellular calcium levels and inhibition of PKC. These findings may explain the reduced cytotoxicity of chemotherapeutic agents in hypoxic/acidic tumors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Extracellular Space/physiology , Hydrogen-Ion Concentration , Calcium/metabolism , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...