Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters










Publication year range
1.
Ann Endocrinol (Paris) ; 71(4): 249-56, 2010 Sep.
Article in French | MEDLINE | ID: mdl-20416860

ABSTRACT

The discovery of apelin, an endogenous ligand of the orphan APJ receptor, constitutes an important advance in both fundamental research and clinical medicine. Experimental data have shown that apelin has a diuretic effect via its central and renal actions: by inhibiting the phasic activity of vasopressinergic neurons and systemic secretion of vasopressin and its direct effect on the renal microcirculation and probably tubular function. Besides its diuretic action, when injected into the blood stream, apelin decreases blood pressure and increases the contractile force of the myocardium while decreasing pre- and post-load, actions opposing those of vasopressin and angiotensin II. Taken together, these data show that this new circulating vasoactive (neuro)peptide could play a crucial role in maintaining water and electrolyte balance and cardiovascular functions. Finally, a systemic injection of apelin in insulin-resistant mice decreases glycemia and enhances glucose uptake in skeletal muscle and adipose tissue, contributing to homeostatic control of blood glucose. Clinically, the development of non-peptide analogs of the apelin receptor could provide new therapeutic tools potentially useful for the treatment of heart failure, states of water and/or electrolyte retention, and type 2 diabetes mellitus.


Subject(s)
Blood Glucose/drug effects , Blood Glucose/physiology , Diuresis , Heart/drug effects , Heart/physiology , Intercellular Signaling Peptides and Proteins/physiology , Water-Electrolyte Balance , Adipose Tissue/drug effects , Amino Acid Sequence , Angiotensin II/antagonists & inhibitors , Animals , Apelin , Blood Pressure/drug effects , Cattle , Diuretics/pharmacology , Humans , Insulin Resistance , Kidney/drug effects , Male , Mice , Molecular Sequence Data , Muscle, Skeletal/drug effects , Rats , Vasopressins/antagonists & inhibitors , Vasopressins/metabolism
2.
J Neuroendocrinol ; 20(3): 279-89, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18194430

ABSTRACT

The hyperactivity of the brain renin-angiotensin system (RAS) has been implicated in the development and maintenance of hypertension in several types of experimental and genetic hypertension animal models. Among the main bioactive peptides of the brain RAS, angiotensin (Ang) II and Ang III display the same affinity for type 1 and type 2 Ang II receptors. Both peptides, injected intracerebroventricularly, similarly increase arginine vasopressin (AVP) release and blood pressure (BP); however, because Ang II is converted in vivo to Ang III, the identity of the true effector is unknown. We review new insights into the predominant role of brain Ang III in the control of BP, underlining the fact that brain aminopeptidase A (APA), the enzyme generating brain Ang III, may therefore be an interesting candidate target for the treatment of hypertension. This justifies the development of potent systemically active APA inhibitors, such as RB150, as prototypes of a new class of antihypertensive agents for the treatment of certain forms of hypertension. We also searched for a putative angiotensin receptor subtype specific for Ang III and isolated a seven transmembrane-domain G protein-coupled receptor corresponding to the receptor for apelin, a newly-discovered peptide isolated from bovine stomach. Apelin and its receptor are expressed in magnocellular vasopressinergic neurones in the hypothalamus. The central injection of apelin in lactating rats decreases the phasic electrical activity of vasopressinergic neurones and the systemic secretion of AVP, inducing water diuresis. Apelin is therefore a natural inhibitor of the antidiuretic effect of AVP. In addition, systemic administration of apelin decreases BP, improves cardiac contractility and reduces cardiac loading. The development of nonpeptide agonists of the apelin receptor may provide new therapeutic tools for treating water retention, hyponatraemia and cardiovascular diseases. Angiotensins and apelin thus exert opposing but complementary effects, and are thereby determinant for the maintenance of body fluid homeostasis and cardiovascular functions.


Subject(s)
Angiotensins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Neurosecretory Systems/physiology , Animals , Apelin , Apelin Receptors , Blood Pressure/drug effects , Brain/metabolism , Brain/physiology , Cardiovascular Physiological Phenomena , Enzyme Inhibitors/pharmacology , Glutamyl Aminopeptidase/antagonists & inhibitors , Humans , Models, Biological , Rats , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Tissue Distribution , Water-Electrolyte Balance/physiology
3.
Arch Mal Coeur Vaiss ; 100(8): 704-8, 2007 Aug.
Article in French | MEDLINE | ID: mdl-17928781

ABSTRACT

Apelin is a peptide involved in the regulation of body fluid homeostasis and cardiovascular functions, that was recently isolated as the endogenous ligand for the human orphan APJ receptor, a G protein-coupled receptor which shares 31% amino-acid sequence identity with the angiotensin II type 1 receptor. The predominant molecular forms of apelin naturally occuring in vivo are apelin 36, apelin 17 (K17F) and the pyroglutamyl form of apelin 13 (pE13F). We investigated the structure-activity relationships of apelin at the rat apelin receptor, tagged at its C-terminal end with enhanced green fluorescent protein and stably expressed in CHO cells. We compared the abilities of N- and C-terminal deleted fragments of K17F (KFRRQRPRLSHKGPMPF) to bind with high affinity to the apelin receptor, to inhibit cAMP production and to induce apelin receptor internalization. The first five N-terminal and the last two C-terminal amino acids of K17F were not essential for apelin binding or cAMP response. In contrast, deletion of the arginine in position 6 drastically decreased binding and cAMP response. The full-length sequence of K17F was the most potent inducer of apelin receptor internalization because successive N-terminal amino-acid deletions progressively reduced internalization and the removal of a single amino acid, the phenylalanine in position 17 at the C-terminus of K17F abolished this process. Thus, K16P binds with high affinity to the apelin receptor and strongly inhibits cAMP production, but does not induce apelin receptor endocytosis. These data indicate that apelin receptor signaling (coupling to Gi) and endocytosis are functionally dissociated, possibly reflecting the existence of several conformational states of this receptor, stabilized by the binding of different apelin fragments to the receptor. We then investigated the consequences for biological activity of this functional dissociation by evaluating the effects of various apelin fragments, injected iv, on arterial blood pressure in normotensive Wistar Kyoto rats. We showed that apelin fragments, that did not induce receptor internalization in vitro but kept their ability to activate receptor coupling to Gi, did not decrease arterial blood pressure. Our data showed that hypotensive actions of apelin peptides correlate with the ability of those ligands to internalize. Thus, the depressor response of apelin may be controlled by apelin receptor endocytosis, which is probably required for initiation of a second wave of signal transduction. The development of biaised agonists of the apelin receptor capable of promoting only one specific signal transduction pathway may therefore offer new therapeutic avenues for the treatment of cardiovascular disorders.


Subject(s)
Blood Pressure/physiology , Carrier Proteins/physiology , Endocytosis/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Apelin , Blood Pressure/drug effects , Carrier Proteins/pharmacology , Intercellular Signaling Peptides and Proteins , Peptide Fragments/pharmacology , Rats , Rats, Inbred WKY , Signal Transduction/physiology , Structure-Activity Relationship
4.
Science ; 315(5813): 766; author reply 766, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17289961

ABSTRACT

Zhang et al. (Research Articles, 11 November 2005, p. 996) reported that obestatin, a peptide derived from the ghrelin precursor, activated the orphan G protein-coupled receptor GPR39. However, we found that I125-obestatin does not bind GPR39 and observed no effects of obestatin on GPR39-transfected cells in various functional assays (cyclic adenosine monophosphate production, calcium mobilization, and GPR39 internalization). Our results indicate that obestatin is not the cognate ligand for GPR39.


Subject(s)
Peptide Hormones/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Calcium/metabolism , Cell Membrane/metabolism , Colforsin/pharmacology , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Ghrelin , Humans , Ligands , Molecular Sequence Data , Peptide Hormones/genetics , Peptide Hormones/pharmacology , Pituitary Gland/cytology , Pituitary Gland/metabolism , Protein Binding , Receptors, G-Protein-Coupled/genetics , Transfection
5.
J Pept Res ; 65(2): 175-88, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15705162

ABSTRACT

Aminopeptidase A (APA) is involved in the maturation of angiotensin III, a peptide which seems to be implicated in blood pressure regulation at the brain level. Therefore APA inhibitors are potential new antihypertensive agents with possible novel applications. With the aim of enhancing the bioavailability and potency of EC 33, the APA inhibitor (Ki = 300 nM) initially used in the earlier studies, we have synthesized new non-peptidic inhibitors able to interact with the S1 and S'1 subsites of the targeted enzyme. Compound 10a, (3S,4S)-3-amino-4-mercapto-6-phenyl-hexane-1-sulfonic acid was obtained using an asymmetric synthesis. Inhibitor 10a exhibits a Ki value of 30 nm.


Subject(s)
Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/pharmacology , Glutamyl Aminopeptidase/antagonists & inhibitors , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/pharmacology , Sulfonic Acids/chemical synthesis , Sulfonic Acids/pharmacology , Animals , Humans , Sulfonic Acids/chemistry
6.
Neuroscience ; 113(3): 653-62, 2002.
Article in English | MEDLINE | ID: mdl-12150785

ABSTRACT

The peptide apelin originating from a larger precursor preproapelin molecule has been recently isolated and identified as the endogenous ligand of the human orphan G protein-coupled receptor, APJ (putative receptor protein related to the angiotensin receptor AT(1)). We have shown recently that apelin and apelin receptor mRNA are expressed in brain and that the centrally injected apelin fragment K17F (Lys(1)-Phe-Arg-Arg-Gln-Arg-Pro-Arg-Leu-Ser-His-Lys-Gly-Pro-Met-Pro-Phe(17)) decreased vasopressin release and altered drinking behavior. Using a specific polyclonal antiserum against K17F for immunohistochemistry, the aim of the present study was to establish the precise topographical distribution of apelin immunoreactivity in colchicine-treated adult rat brain. Immunoreactivity was essentially detected in neuronal cell bodies and fibers throughout the entire neuroaxis in different densities. Cells bodies have been visualized in the preoptic region, the hypothalamic supraoptic and paraventricular nuclei and in the highest density, in the arcuate nucleus. Apelin immunoreactive cell bodies were also seen in the pons and the medulla oblongata. Apelin nerve fibers appear more widely distributed than neuronal apelin cell bodies. The hypothalamus represented, by far, the major site of apelin-positive nerve fibers which were found in the suprachiasmatic, periventricular, dorsomedial, ventromedial nuclei and in the retrochiasmatic area, with the highest density in the internal layer of the median eminence. Fibers were also found innervating other circumventricular organs such as the vascular organ of the lamina terminalis, the subfornical and the subcommissural organs and the area postrema. Apelin was also detected in the septum and the amygdala and in high density in the paraventricular thalamic nucleus, the periaqueductal central gray matter and dorsal raphe nucleus, the parabrachial and Barrington nuclei in the pons and in the nucleus of the solitary tract, lateral reticular, prepositus hypoglossal and spinal trigeminal nuclei. The topographical distribution of apelinergic neurons in the brain suggests multiple roles for apelin especially in the central control of ingestive behaviors, pituitary hormone release and circadian rhythms.


Subject(s)
Brain/cytology , Carrier Proteins/analysis , Neurons/chemistry , Animals , Apelin , Brain Chemistry , Carrier Proteins/biosynthesis , Carrier Proteins/immunology , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Male , Neurons/metabolism , Rats , Rats, Inbred WKY
7.
J Comp Neurol ; 440(2): 192-203, 2001 Nov 12.
Article in English | MEDLINE | ID: mdl-11745617

ABSTRACT

Studies have demonstrated a specific function of the angiotensin II (Ang II) type 1 receptor (AT(1)) in regulation of adult central cardiovascular, fluid, and pituitary hormone release and a predominant role of the renin-angiotensin system in fetal and neonatal cardiovascular homeostasis. The pattern of brain AT(1) mRNA expression during fetal and neonatal development is currently unknown. We used radiolabeled cRNA probes for in situ hybridization histochemistry to determine the ontogenic development of the two AT(1) subtypes (AT(1a) and AT(1b)) mRNA in rat brain, from 11 days of gestation (E11) to 28 days after birth (P28). No AT(1b) mRNA was detected in the developing brain, whereas AT(1a) mRNA was first detected at E19. The age at which AT(1a) mRNA is first detected varied among different brain areas and expression predominates in areas involved in fluid homeostasis, pituitary hormone release, and cardiovascular regulation, where it persists until P28. AT(1a) mRNA expression is present from E19 onward in the median preoptic nucleus, the vascular organ of the lamina terminalis, the paraventricular nucleus, the periaqueductal gray, the nucleus raphe pallidus, the motor facial nucleus, and very weakly in the nucleus of the solitary tract and the ambiguous nucleus, and at E21 in the subfornical organ, the anterior olfactory nucleus and the piriform cortex. AT(1a) mRNA expression is present after birth in many regions, including the preoptic and lateral hypothalamic areas, the area postrema and medullary reticular nuclei. In conclusion, during brain development, expression of AT(1a) mRNA, appears in late gestation at E19, predominantly in forebrain areas involved in fluid homeostasis and cardiovascular regulation. In contrast, AT(1a) mRNA expression is absent or present only in very small amounts until after birth in many medullary nuclei, known to play an important role in cardiovascular modulation. Our results suggest that, in perinatal life, AT(1a) is involved in fluid and perhaps cardiovascular homeostasis and that the role of Ang II in modulating medullary cardiovascular centers matures later in postnatal life.


Subject(s)
Angiotensin II/metabolism , Brain/embryology , Cardiovascular Physiological Phenomena , Homeostasis/genetics , Neurons/metabolism , Receptors, Angiotensin/genetics , Aging/genetics , Animals , Animals, Newborn , Brain/growth & development , Brain/metabolism , Diencephalon/embryology , Diencephalon/growth & development , Diencephalon/metabolism , Female , Fetus , In Situ Hybridization , Male , Mesencephalon/embryology , Mesencephalon/growth & development , Mesencephalon/metabolism , Neurons/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Rhombencephalon/embryology , Rhombencephalon/growth & development , Rhombencephalon/metabolism , Telencephalon/embryology , Telencephalon/growth & development , Telencephalon/metabolism , Water-Electrolyte Balance/genetics
8.
Biochemistry ; 40(48): 14440-8, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11724556

ABSTRACT

Aminopeptidase A (EC 3.4.11.7, APA) is a 160 kDa membrane-bound zinc enzyme that contains the HEXXH consensus sequence found in members of the zinc metalloprotease family, the zincins. In addition, the monozinc aminopeptidases are characterized by another conserved motif, GXMEN, the glutamate residue of which has been shown to be implicated in the exopeptidase specificity of aminopeptidase A [Vazeux G. (1998) Biochem. J. 334, 407-413]. In carboxypeptidase A (EC 3.4.17.1, CPA), the exopeptidase specificity is conferred by an arginine residue (Arg-145) and an asparagine residue (Asn-144). Thus, we hypothesized that Asn-353 of the GXMEN motif in APA plays a similar role to Asn-144 in CPA and contributes to the exopeptidase specificity of APA. We investigated the functional role of Asn-353 in APA by substituting this residue with a glutamine (Gln-353), an alanine (Ala-353) or an aspartate (Asp-353) residue by site-directed mutagenesis. Expression of wild-type and mutated APAs revealed that Gln-353 and Ala-353 are similarly routed and glycosylated to the wild-type APA, whereas Asp-353 is trapped intracellularly and partially glycosylated. Kinetic studies, using alpha-L-glutamyl-beta-naphthylamide (GluNA) as a substrate showed that the K(m) values of the mutants Gln-353 and Ala-353 were increased 11- and 8-fold, respectively, whereas the k(cat) values were decreased (2-fold) resulting in a 24- and 14-fold reduction in cleavage efficiency. When alpha-L-aspartyl-beta-naphthylamide or angiotensin II were used as substrates, the mutations had a greater effect on k(cat), leading to a similar decrease in cleavage efficiencies as that observed with GluNA. We then measured the inhibitory potencies of several classes of inhibitors, glutamate thiol, glutamine thiol and two isomers (L- or D-) of glutamate phosphonate to explore the functional role of Asn-353. The data indicate that Asn-353 is critical for the integrity and catalytic activity of APA. This residue is involved in substrate binding via interactions with the free N-terminal part and with the P1 carboxylate side chain of the substrate. In conclusion, Asn-353 of the GXMEN motif, together with Glu-352, contributes to the exopeptidase specificity of APA and plays an equivalent role to Asn-144 in CPA.


Subject(s)
Aminopeptidases/chemistry , Asparagine/chemistry , Metalloendopeptidases/chemistry , Zinc/chemistry , Amino Acid Sequence , Angiotensin II/metabolism , Animals , CHO Cells , Consensus Sequence , Cricetinae , DNA Primers , Escherichia coli/enzymology , Fluorescent Antibody Technique , Glutamic Acid/chemistry , Glutamyl Aminopeptidase , Humans , Hydrolysis , Kinetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Precipitin Tests , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
9.
Endocrinology ; 142(11): 4683-92, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606433

ABSTRACT

Differences involving serine residues in the sequence of the carboxyl-terminal tail of type 1 angiotensin II (Ang II) receptor subtypes AT(1A) and AT(1B) suggest differences in desensitization ability. We examined the Ang II-induced homologous desensitization patterns of both receptor subtypes in freshly isolated renal structures: glomerulus (Glom), afferent arteriole, and cortical thick ascending limb (CTAL), whose content in each subtype mRNA is different, by measuring variations in intracellular calcium concentration. A preexposure to a maximal dose of Ang II, followed by a second application of the same concentration, induced: 1) a complete desensitization in Glom, where AT(1A) and AT(1B) mRNAs were expressed in similar proportions, and 2) no or partial desensitization in afferent arteriole and CTAL, where AT(1A) mRNA was predominant. In the absence of nephron structure containing only AT(1B) mRNA, we studied rat anterior pituitary cells that exhibit high content in this subtype and observed that desensitization was not complete. In Glom, CTAL, and pituitary cells, desensitization proceeded in a dose-dependent manner. In Glom and CTAL, desensitization occurred via a PKC-independent mechanism. These results suggest that desensitization does not depend on the nature of Ang II receptor subtype but either on the proportion of each subtype in a given cell and/or on cell specific type. This could allow adaptive biological responses to Ang II appropriate to the specific function of a given cell type.


Subject(s)
Angiotensin II/pharmacology , Kidney/metabolism , Receptors, Angiotensin/metabolism , Animals , Arterioles/metabolism , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Intracellular Membranes/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Loop of Henle/cytology , Loop of Henle/metabolism , Male , Osmolar Concentration , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/drug effects
10.
J Neurochem ; 77(4): 1085-96, 2001 May.
Article in English | MEDLINE | ID: mdl-11359874

ABSTRACT

Apelin, a peptide recently isolated from bovine stomach tissue extracts, has been identified as the endogenous ligand of the human orphan APJ receptor. We established a stable Chinese hamster ovary (CHO) cell line expressing a gene encoding the rat apelin receptor fused to the enhanced green fluorescent protein, to investigate internalization and the pharmacological profile of the apelin receptor. Stimulation of this receptor by the apelin fragments K17F (Lys1-Phe-Arg-Arg-Gln-Arg-Pro-Arg-Leu-Ser-His-Lys-Gly-Pro-Met-Pro-Phe17) and pE13F (pGlu5-Arg-Pro-Arg-Leu-Ser-His-Lys-Gly-Pro-Met-Pro-Phe17) resulted in a dose-dependent inhibition of forskolin-induced cAMP production and promoted its internalization. In contrast, the apelin fragments R10F (Arg8-Leu-Ser-His-Lys-Gly-Pro-Met-Pro-Phe17) and G5F (Gly13-Pro-Met-Pro-Phe17) were inactive. The physiological role of apelin and its receptor was then investigated by showing for the first time in rodent brain: (i) detection of apelin neurons in the supraoptic and paraventricular nuclei by immunohistochemistry with a specific polyclonal anti-apelin K17F antibody; (ii) detection of apelin receptor mRNA in supraoptic vasopressinergic neurons by in situ hybridization and immunohistochemistry; and (iii) a decrease in vasopressin release following intracerebroventricular injection of K17F, or pE13F, but not R10F. Thus, apelin locally synthesized in the supraoptic nucleus could exert a direct inhibitory action on vasopressinergic neuron activity via the apelin receptors synthesized in these cells. Furthermore, central injection of pE13F significantly decreased water intake in dehydrated normotensive rats but did not affect blood pressure. Together, these results suggest that neuronal apelin plays an important role in the central control of body fluid homeostasis.


Subject(s)
Brain/physiology , Carrier Proteins/physiology , Receptors, Dopamine D2/physiology , Receptors, G-Protein-Coupled , Adipokines , Amino Acid Sequence , Animals , Apelin , Apelin Receptors , Blood Pressure/drug effects , CHO Cells , Carrier Proteins/chemistry , Carrier Proteins/pharmacology , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Drinking Behavior/drug effects , In Situ Hybridization , Injections, Intravenous , Injections, Intraventricular , Intercellular Signaling Peptides and Proteins , Male , Mice , Molecular Sequence Data , Neurons/physiology , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , RNA, Messenger/analysis , Rats , Rats, Inbred WKY , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/genetics , Transfection , Vasopressins/blood , Water Deprivation
11.
Trends Endocrinol Metab ; 12(4): 157-62, 2001.
Article in English | MEDLINE | ID: mdl-11295571

ABSTRACT

Among the main bioactive peptides of the brain renin-angiotensin system, angiotensin (Ang) II and AngIII exhibit the same affinity for type 1 and type 2 AngII receptors. Both peptides, injected intracerebroventricularly, cause similar increases in vasopressin release and blood pressure. Because AngII is converted in vivo to AngIII, the identity of the true effector is unknown. This review summarizes new insights into the predominant role of brain AngIII in the control of vasopressin release and blood pressure and underlines the fact that brain aminopeptidase A, the enzyme forming central AngIII, could constitute a putative central therapeutic target for the treatment of hypertension.


Subject(s)
Angiotensin III/physiology , Blood Pressure , Vasopressins/metabolism , Angiotensin II/administration & dosage , Angiotensin II/physiology , Angiotensin III/administration & dosage , Animals , Arginine Vasopressin/metabolism , Brain/drug effects , Brain/physiology , Humans , Injections, Intraventricular
12.
J Histochem Cytochem ; 48(11): 1553-64, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11036098

ABSTRACT

Because G-protein-coupled receptors (GPCRs) constitute excellent putative therapeutic targets, functional characterization of orphan GPCRs through identification of their endogenous ligands has great potential for drug discovery. We propose here a novel single cell-based assay for identification of these ligands. This assay involves (a) fluorescent tagging of the GPCR, (b) expression of the tagged receptor in a heterologous expression system, (c) incubation of the transfected cells with fractions purified from tissue extracts, and (d) imaging of ligand-induced receptor internalization by confocal microscopy coupled to digital image quantification. We tested this approach in CHO cells stably expressing the NT1 neurotensin receptor fused to EGFP (enhanced green fluorescent protein), in which neurotensin promoted internalization of the NT1-EGFP receptor in a dose-dependent fashion (EC(50) = 0.98 nM). Similarly, four of 120 consecutive reversed-phase HPLC fractions of frog brain extracts promoted internalization of the NT1-EGFP receptor. The same four fractions selectively contained neurotensin, an endogenous ligand of the NT1 receptor, as detected by radioimmunoassay and inositol phosphate production. The present internalization assay provides a highly specific quantitative cytosensor technique with sensitivity in the nanomolar range that should prove useful for the identification of putative natural and synthetic ligands for GPCRs.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Neurotensin/metabolism , Tissue Extracts/metabolism , Animals , Brain/metabolism , CHO Cells , Chromatography, High Pressure Liquid , Cricetinae , Green Fluorescent Proteins , Image Processing, Computer-Assisted , Inositol Phosphates/biosynthesis , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Neurotensin/pharmacology , Radioimmunoassay , Radioligand Assay , Rana ridibunda , Receptors, Neurotensin/genetics , Recombinant Fusion Proteins/metabolism , Sensitivity and Specificity , Transfection
13.
Biochem Soc Trans ; 28(4): 435-40, 2000.
Article in English | MEDLINE | ID: mdl-10961935

ABSTRACT

Overactivity of the brain renin-angiotensin system (RAS) has been implicated in the development and maintenance of hypertension in several experimental animal models. We have recently reported that, in the murine brain RAS, angiotensin II (AngII) is converted by aminopeptidase A (APA) into angiotensin III (AngIII),which is itself degraded by aminopeptidase N (APN), both peptides being equipotent to increase vasopressin release and arterial blood pressure when injected by the intracerebroventricular (i.c.v.) route. Because AngII is converted in vivo into AngIII, the exact nature of the active peptide is not precisely known. To delineate their respective roles in the central control of cardiovascular functions, specific and selective APA and APN inhibitors are needed to block the metabolic pathways of AngII and AngIII respectively. In the absence of such compounds for APA, we first explored the organization of the APA active site by site-directed mutagenesis. This led us to propose a molecular mechanism of action for APA similar to that proposed for the bacterial enzyme thermolysin deduced from X-ray diffraction studies. Secondly, we developed a specific and selective APA inhibitor, compound EC33 [(S)-3-amino-4-mercaptobutylsulphonic acid], as well as a potent and selective APN inhibitor, PC18 (2-amino-4-methylsulphonylbutane thiol). With these new tools we examined the respective roles of AngII and AngIII in the central control of arterial blood pressure. A central blockade of APA with the APA inhibitor EC33 suppressed the pressor effect of exogenous AngII, suggesting that brain AngII must be converted into AngIII to increase arterial blood pressure. Furthermore, EC33, injected alone i.c.v. but not intravenously, caused a dose-dependent decrease in arterial blood pressure by blocking the formation of brain AngIII but not systemic AngIII. This is corroborated by the fact that the selective APN inhibitor PC18 administered alone via the i.c.v. route increased arterial blood pressure. This pressor response was blocked by prior treatment with the angiotensin type 1 receptor antagonist losartan, showing that blocking the action of APN on AngIII metabolism leads to an increase in endogenous AngIII levels, resulting in arterial blood pressure increase through an interaction with angiotensin type 1 receptors. These results demonstrate that AngIII is a major effector peptide of the brain RAS, exerting a tonic stimulatory control over arterial blood pressure. Thus APA, the enzyme responsible for the formation of brain AngIII, represents a potential central therapeutic target that justifies the development of APA inhibitors, crossing the blood-brain barrier, as central anti-hypertensive agents.


Subject(s)
Aminopeptidases/physiology , Angiotensin III/biosynthesis , Arteries/physiology , Blood Pressure , Brain/metabolism , Renin-Angiotensin System , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Angiotensin II/metabolism , Angiotensin Receptor Antagonists , Animals , Antihypertensive Agents/pharmacology , Binding Sites , CD13 Antigens/metabolism , Dose-Response Relationship, Drug , Glutamyl Aminopeptidase , Hypertension/drug therapy , Hypothalamus/metabolism , Losartan/pharmacology , Mice , Models, Chemical , Mutagenesis, Site-Directed , Peptides/metabolism , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Thermolysin/metabolism , Vasopressins/metabolism
14.
Biochemistry ; 39(11): 3061-8, 2000 Mar 21.
Article in English | MEDLINE | ID: mdl-10715127

ABSTRACT

Aminopeptidase A (EC 3.4.11.7, APA) is a 130 kDa membrane-bound protease that contains the HEXXH consensus sequence found in the zinc metalloprotease family, the zincins. In addition to the catalytic zinc atom, APA contains a Ca2+ ion that increases its enzymatic activity. Aligning the sequences of the mouse APA, APN, and other monozinc aminopeptidases led to the identification of a conserved histidine (His 450 in mouse APA). Replacing this residue with a phenylalanine (Phe 450) by site-directed mutagenesis resulted in markedly lower levels of APA activity and in a change in the sensitivity of APA to Ca2+ (the EC50 for Ca2+ was 25 microM in the wild type and only 279 microM in the mutant). Kinetic studies, with a supramaximal Ca2+ concentration (4 mM), showed that the Km of the mutant enzyme for the substrate alpha-L-glutamyl-beta-naphthylamide was 25 times higher than that of the wild type, whereas the kcat value was much lower (factor of 22). Thus, overall, the wild-type enzyme had a cleavage efficiency that was 571 times higher than that of the mutant. The inhibitory potencies of two different classes of inhibitors, a glutamate thiol and a glutamate phosphonate compound, were significantly lower (factors of 19 and 22, respectively) for the mutated enzyme than for the wild-type enzyme. In contrast, inhibition by lysine thiol was unaffected. These data strongly suggest that His 450 is critical for catalytic activity and is involved in substrate binding via interaction with the P1 carboxylate side chain of the substrate. Furthermore, His 450, together with Ca2+, may contribute to the substrate specificity of APA for N-terminal acidic amino acid residues.


Subject(s)
Aminopeptidases/metabolism , Calcium/metabolism , Histidine/metabolism , Amino Acid Sequence , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/genetics , Aminopeptidases/standards , Animals , Asparagine/genetics , CHO Cells , Calcium/physiology , Catalysis , Cricetinae , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Glutamates/pharmacology , Glutamyl Aminopeptidase , Histidine/genetics , Humans , Lysine/pharmacology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Organophosphonates/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/standards , Substrate Specificity/genetics , Sulfhydryl Compounds/pharmacology
15.
Biochemistry ; 39(5): 1152-5, 2000 Feb 08.
Article in English | MEDLINE | ID: mdl-10653662

ABSTRACT

Through the development of a new chemical strategy, aminophosphinic peptides containing a pseudoglutamyl residue (Glu Psi(PO2-CH2)Leu-Xaa) in the N-terminal position were synthesized and evaluated as inhibitors of aminopeptidase A (APA). The most potent inhibitor developed in this study, Glu Psi(PO2-CH2)Leu-Ala, displayed a Ki value of 0.8 nM for APA, but was much less effective in blocking aminopeptidase N (APN) (Ki = 31 microM). The critical role of the glutamyl residue in this phosphinic peptide, both in potency and selectivity, is exemplified by the P1 position analogue, Ala Psi(PO2-CH2)Leu-Ala, which exhibited a Ki value of 0.9 microM toward APA but behaved as a rather potent inhibitor of APN (Ki = 25 nM). Glu Psi(PO2-CH2)Leu-Xaa peptides are poor inhibitors of angiotensin converting enzyme (Ki values higher than 1 microM). Depending on the nature of the Xaa residue, the potency of these phosphinic peptides toward neutral endopeptidase 24-11 varied from 50 nM to 3 microM. In view of the in vivo role of APA in the formation of brain angiotensin III, one of the main effector peptides of the renin angiotensin system in the central nervous system, highly potent and selective inhibitors of APA may find important therapeutic applications soon.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Aminopeptidases/chemistry , Enzyme Inhibitors/chemistry , Glutamic Acid/chemistry , Peptides/chemistry , Phosphinic Acids/chemistry , Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , CD13 Antigens/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glutamic Acid/chemical synthesis , Glutamyl Aminopeptidase , Humans , Neprilysin/antagonists & inhibitors , Peptides/chemical synthesis , Peptides/pharmacology , Phosphinic Acids/pharmacology , Rabbits , Zinc/chemistry
16.
Neuroendocrinology ; 72(6): 400-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11146423

ABSTRACT

The peptide apelin, recently isolated from bovine stomach tissue extracts, has been identified as an endogenous ligand of the human putative receptor protein related to the angiotensin receptor AT(1) (APJ). In this article, we report cloning of the rat apelin receptor cDNA. The sequence shares 90% identity with the human APJ receptor and 31% with the rat AT(1A) angiotensin receptor. Subsequently a stable CHO cell line expressing the receptor fused at its C-terminal part with the enhanced green fluorescent protein (EGFP) was established, allowing to verify its cell surface distribution and to determine the affinity of various apelin and angiotensin fragments on the cloned receptor. As shown for the human APJ receptor, the rat apelin receptor expressed in the cell line was negatively coupled to adenylate cyclase. The apelin fragment K17F (Lys(1)-Phe-Arg-Arg-Gln-Arg-Pro-Arg-Leu-Ser-His-Lys-Gly-Pro-Met-Pro-Phe(17)) inhibited forskolin-stimulated cAMP production at sub-nanomolar concentrations whereas angiotensin II and angiotensin III were inactive. N-terminal elongation of K17F with a tyrosine or the N-terminal deletion of the first four amino acids did not modify the inhibitory action of K17F on cAMP production. In contrast, deletion of the first seven amino acids of K17F or substitution of phenylalanine by an alanine residue at the C-terminus completely abolished the activity of the peptide. In situ hybridization analysis of apelin receptor mRNA expression in the adult rat brain showed intense labeling in the hypothalamus, especially in the supraoptic and the paraventricular nuclei. The anterior and intermediate lobes of the pituitary were also highly labeled, as well as the pineal gland. Labeling was also found in extrahypothalamic structures such as the piriform cortex, the nucleus of the lateral olfactory tract, the central grey matter, the pars compacta of the substantia nigra, the dorsal raphe nucleus, the entorhinal cortex, the dentate gyrus and the Ammon's horn. The hypothalamic and hypophyseal distribution of the receptor suggests an involvement of apelin in the control of neuro- and adenohypophyseal hormone release, whereas its presence in the pineal gland and in discrete higher brain structures points out to possible roles in the regulation of circadian rhythms and of water and food intake behavior.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled , Amino Acid Sequence , Angiotensins/pharmacology , Animals , Apelin , Apelin Receptors , Base Sequence , CHO Cells , Carrier Proteins/pharmacology , Circadian Rhythm/physiology , Cloning, Molecular , Colforsin/pharmacology , Cricetinae , Cyclic AMP/metabolism , Gene Expression/physiology , Humans , Hypothalamus, Anterior/chemistry , Hypothalamus, Anterior/physiology , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Molecular Sequence Data , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/physiology , Pituitary Gland/chemistry , Pituitary Gland/physiology , RNA, Messenger/analysis , Rats , Receptors, Angiotensin/metabolism , Transfection
17.
Eur J Pharmacol ; 384(2-3): 223-30, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10611445

ABSTRACT

Rat mesangial cells were exposed to angiotensin II, angiotensin AT(1) receptor antagonists such as losartan, EXP 3174 and candesartan, or dexamethasone for increasing periods (1-24 h). Angiotensin AT(1A) and AT(1B) receptor mRNA were measured by reverse transcription-polymerase chain reaction (RT-PCR). Angiotensin II, losartan and EXP 3174 did not modify significantly angiotensin AT(1A) and AT(1B) receptor mRNA. Candesartan increased angiotensin AT(1B) receptor mRNA and, to a lesser extent, angiotensin AT(1A) receptor mRNA. In contrast, dexamethasone decreased mainly angiotensin AT(1B) receptor mRNA. As shown by Western blot analysis, exposure of mesangial cells to angiotensin II, losartan or EXP 3174 did not produce any change in angiotensin AT(1) receptor protein, whereas dexamethasone and candesartan exerted inhibitory effects. In conclusion, the angiotensin AT(1B) receptor subtype, the most abundantly distributed in rat mesangial cells, is inhibited by glucocorticoids. The effect of candesartan is more complex with a slight stimulation of angiotensin AT(1B) mRNA and a marked inhibition of angiotensin AT(1) receptor protein. In contrast, angiotensin II and the other angiotensin AT(1) receptor antagonists studied are inactive on angiotensin AT(1) mRNA and protein.


Subject(s)
Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Glomerular Mesangium/drug effects , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Imidazoles/pharmacology , Losartan/pharmacology , Male , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Receptors, Angiotensin/metabolism , Tetrazoles/pharmacology
18.
J Med Chem ; 42(25): 5197-211, 1999 Dec 16.
Article in English | MEDLINE | ID: mdl-10602705

ABSTRACT

The study of the physiological roles of the membrane-bound zinc-aminopeptidase A (glutamyl aminopeptidase, EC 3.4.11.7) needs the design of efficient and selective inhibitors of this enzyme. An acute exploration of aminopeptidase A active site was performed by a combinatorial approach using (3-amino-2-mercapto-acyl)dipeptides able to fit its S(1), S(1)', and S(2)' subsites. This analysis confirmed that the S(1) subsite is optimally blocked by a glutamate or isosteric residues and demonstrated that the S(1)' subsite is hydrophobic whereas the S(2)' subsite recognizes preferentially negatively charged residues derived from aspartic acid. The optimization of these structural parameters led to the synthesis of nanomolar and subnanomolar inhibitors of aminopeptidase A such as H(3)N(+)CH(CH(2)CH(2)SO(3)(-))CH(SH)CO-Ile-(3-COOH)Pro that exhibits a K(i) of 0.87 nM. The best compounds were synthesized by a stereochemically controlled route. These first described highly potent inhibitors could allow studies about the role of physiological substrates of APA such as angiotensin II and cholecystokinin CCK(8) in the central nervous system.


Subject(s)
Aminopeptidases/metabolism , Enzyme Inhibitors/metabolism , Aminopeptidases/antagonists & inhibitors , Animals , CD13 Antigens/antagonists & inhibitors , CD13 Antigens/metabolism , Enzyme Inhibitors/chemistry , Glutamyl Aminopeptidase , Magnetic Resonance Spectroscopy , Molecular Structure , Neprilysin/metabolism , Peptidyl-Dipeptidase A/drug effects , Peptidyl-Dipeptidase A/metabolism , Rabbits , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Stereoisomerism , Swine
19.
Proc Natl Acad Sci U S A ; 96(23): 13415-20, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10557335

ABSTRACT

Overactivity of the brain renin-angiotensin system (RAS) has been implicated in the development and maintenance of hypertension in several experimental models, such as spontaneously hypertensive rats and transgenic mice expressing both human renin and human angiotensinogen transgenes. We recently reported that, in the murine brain, angiotensin II (AngII) is converted to angiotensin III (AngIII) by aminopeptidase A (APA), whereas AngIII is inactivated by aminopeptidase N (APN). If injected into cerebral ventricles (ICV), AngII and AngIII cause similar pressor responses. Because AngII is metabolized in vivo into AngIII, the exact nature of the active peptide is not precisely determined. Here we report that, in rats, ICV injection of the selective APA inhibitor EC33 [(S)-3-amino-4-mercaptobutyl sulfonic acid] blocked the pressor response of exogenous AngII, suggesting that the conversion of AngII to AngIII is required to increase blood pressure (BP). Furthermore, ICV injection, but not i.v. injection, of EC33 alone caused a dose-dependent decrease in BP by blocking the formation of brain but not systemic AngIII. This is corroborated by the fact that the selective APN inhibitor, PC18 (2-amino-4-methylsulfonyl butane thiol), administered alone via the ICV route, increases BP. This pressor response was blocked by prior treatment with the angiotensin type 1 (AT(1)) receptor antagonist, losartan, showing that blocking the action of APN on AngIII metabolism leads to an increase in endogenous AngIII levels, resulting in BP increase, through interaction with AT(1) receptors. These data demonstrate that AngIII is a major effector peptide of the brain RAS, exerting tonic stimulatory control over BP. Thus, APA, the enzyme responsible for the formation of brain AngIII, represents a potential central therapeutic target that justifies the development of APA inhibitors as central antihypertensive agents.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antihypertensive Agents/pharmacology , Brain/drug effects , Enzyme Inhibitors/pharmacology , Angiotensin II/metabolism , Angiotensin III/metabolism , Animals , Blood Pressure/drug effects , Brain/metabolism , Glutamyl Aminopeptidase , Injections, Intravenous , Injections, Intraventricular , Male , Rats , Rats, Inbred SHR
20.
Kidney Int ; 56(3): 850-9, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10469354

ABSTRACT

BACKGROUND: This study examined the specific effects of angiotensin III (Ang III) along the nephron. METHODS: We examined the distribution of aminopeptidase A (APA) activity by using a specific APA inhibitor and by immunostaining with an antirat kidney APA antibody, the Ang III-induced variations of [Ca2+]i by using fura-2 and the characterization of the receptor subtype involved in the response to Ang III in cortical thick ascending limb (CTAL). RESULTS: APA activity was found all along the nephron but was higher in the cortex than in the medulla. This was confirmed by immunostaining. Increases in [Ca2+]i elicited by 10(-7) mol/liter Ang III were observed all along the nephron. The characterization of the receptor subtype involved in the [Ca2+]i response to Ang III in CTAL indicated that EC50 values for Ang III and Ang II were similar (13.5 and 10.3 nmol/liter, respectively), and Ang III-induced responses were totally abolished by AT1 receptor but not by AT2 receptor antagonists. There was a cross-desensitization of [Ca2+]i responses to 10(-7) mol/liter Ang III and Ang II, and the [Ca2+]i responses to 10(-7) mol/liter Ang II and Ang III were not additive. CONCLUSION: These results show that in CTAL, the [Ca2+]i responses to Ang II and Ang III occur through the same AT1a receptor because this subtype is predominant in this segment. Taken together, these data suggest that APA could be a key enzyme to generate Ang III from Ang II in the kidney.


Subject(s)
Aminopeptidases/metabolism , Angiotensin III/pharmacology , Calcium/metabolism , Nephrons/drug effects , Nephrons/metabolism , Angiotensin II/analogs & derivatives , Angiotensin II/metabolism , Angiotensin II/pharmacology , Angiotensin III/biosynthesis , Angiotensin Receptor Antagonists , Animals , Glutamyl Aminopeptidase , Imidazoles/pharmacology , Immunohistochemistry , In Vitro Techniques , Intracellular Fluid/metabolism , Losartan/pharmacology , Male , Nephrons/enzymology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/drug effects , Receptors, Angiotensin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...