Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Chemistry ; 30(35): e202401296, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38641990

RESUMO

To fill the need for environmentally sensitive fluorescent unnatural amino acids able to operate in the red region of the spectrum, we have designed and synthesized Alared, a red solvatochromic and fluorogenic amino acid derived from the Nile Red chromophore. The new unnatural amino acid can be easily integrated into bioactive peptides using classical solid-phase peptide synthesis. The fluorescence quantum yield and the emission maximum of Alared-labeled peptides vary in a broad range depending on the peptide's environment, making Alared a powerful reporter of biomolecular interactions. Due to its red-shifted absorption and emission spectra, Alared-labeled peptides could be followed in living cells with minimal interference from cellular autofluorescence. Using ratiometric fluorescence microscopy, we were able to track the fate of the Alared-labeled peptide agonists of the apelin G protein-coupled receptor upon receptor activation and internalization. Due to its color-shifting environmentally sensitive emission, Alared allowed for distinguishing the fractions of peptides that are specifically bound to the receptor or unspecifically bound to different cellular membranes.


Assuntos
Aminoácidos , Corantes Fluorescentes , Microscopia de Fluorescência , Peptídeos , Corantes Fluorescentes/química , Peptídeos/química , Aminoácidos/química , Humanos , Microscopia de Fluorescência/métodos , Oxazinas/química , Técnicas de Síntese em Fase Sólida , Espectrometria de Fluorescência
2.
Bioconjug Chem ; 34(1): 162-168, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36534753

RESUMO

Herein, we describe a catalyst-free thia-Diels-Alder cycloaddition for the chemoselective labeling of fully deprotected phosphonodithioester-peptides in solution with fluorophores functionalized with an exocyclic diene. The reaction was optimized on the model tripeptide 1 containing a lysine residue, which enabled its rapid and straightforward labeling with three different fluorophores (fluorescein, lissamine rhodamine B, and squaraine) in very mild conditions (H2O/iPrOH, 37 °C, 1 h). The reaction was then successfully applied to the chemoselective labeling of fully deprotected apelin-13 with squaraine dye. The resulting fluorescent ligand 18 exhibited a high affinity (0.17 ± 0.03 nM) for apelinR. It enabled the development of time-resolved FRET-based competition assays for high-throughput screening and drug discovery. Thanks to its fluorogenic properties, ligand 18 was also successfully involved in the live-cell optical imaging of apelinR in no-wash conditions.


Assuntos
Corantes Fluorescentes , Peptídeos , Apelina , Reação de Cicloadição , Ligantes , Peptídeos/química , Corantes Fluorescentes/química
3.
Int J Mol Sci ; 24(3)2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36768653

RESUMO

The renin-angiotensin system (RAS) is one of the main regulatory systems of cardiovascular homeostasis. It is mainly composed of angiotensin-converting enzyme (ACE) and angiotensin II receptors AT1 and AT2. ACE and AT1 are targets of choice for the treatment of hypertension, whereas the AT2 receptor is still not exploited due to the lack of knowledge of its physiological properties. Peptide toxins from venoms display multiple biological functions associated with varied chemical and structural properties. If Brazilian viper toxins have been described to inhibit ACE, no animal toxin is known to act on AT1/AT2 receptors. We screened a library of toxins on angiotensin II receptors with a radioligand competition binding assay. Functional characterization of the selected toxin was conducted by measuring second messenger production, G-protein activation and ß-arrestin 2 recruitment using bioluminescence resonance energy transfer (BRET) based biosensors. We identified one original toxin, A-CTX-cMila, which is a 7-residues cyclic peptide from Conus miliaris with no homology sequence with known angiotensin peptides nor identified toxins, displaying a 100-fold selectivity for AT1 over AT2. This toxin shows a competitive antagonism mode of action on AT1, blocking Gαq, Gαi3, GαoA, ß-arrestin 2 pathways and ERK1/2 activation. These results describe the first animal toxin active on angiotensin II receptors.


Assuntos
Hipertensão , Receptor Tipo 1 de Angiotensina , Humanos , Angiotensina II/metabolismo , Antagonistas de Receptores de Angiotensina , beta-Arrestina 2/metabolismo , Peptídeos/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores de Angiotensina/metabolismo , Sistema Renina-Angiotensina/fisiologia , Animais
4.
FASEB J ; 34(6): 7989-8000, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32301550

RESUMO

Like apelin (pE13F, K17F), Elabela/Toddler is an endogenous ligand of the apelin receptor playing a key role in cardiovascular development. Elabela/Toddler exists as peptide fragments of 32 (Q32P), 22 (K22P) and 11 (C11P) amino acids. In this study, we investigated the possible structural and functional similarities between these endogenous ligands. We performed in vitro pharmacological characterization and biased signaling analyses for apelin and Elabela/Toddler fragments in CHO cells, by assessing binding affinities, the inhibition of cyclic adenosine monophosphate (cAMP) production and the triggering of ß-arrestin 2 recruitment. We also performed Alanine scanning for Elabela/Toddler and structure-function studies based on site-directed mutagenesis of the rat and human apelin receptor, to compare the modes of binding of the different endogenous ligands. Alanine scanning of K22P showed that neither of its cysteine residues were involved in binding or in peptide activity and that its C-terminus carried the key pharmacophore for receptor binding and activation. We showed that Asp282 and Asp284 of rat and human apelin receptor, respectively, were not involved in Elabela/Toddler activity, whereas they are key residues for apelin binding and activity. We found that the structural features of Elabela/Toddler and apelin were different, resulting in different modes of binding of these endogenous ligands to the apelin receptor. These differences should be taken into account in the future development metabolically stable analogs of Elabela/Toddler and apelin as potential therapeutic tools for the treatment of cardiovascular diseases and water retention/hyponatremic disorders.


Assuntos
Receptores de Apelina/metabolismo , Hormônios Peptídicos/metabolismo , Animais , Células CHO , Doenças Cardiovasculares/metabolismo , Linhagem Celular , Cricetulus , AMP Cíclico/metabolismo , Cisteína/metabolismo , Humanos , Hiponatremia/metabolismo , Fragmentos de Peptídeos/metabolismo , Ratos , Transdução de Sinais/fisiologia
5.
Biochem J ; 477(21): 4133-4148, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-32955085

RESUMO

Aminopeptidase A (APA) is a membrane-bound monozinc aminopeptidase. In the brain, APA generates angiotensin III which exerts a tonic stimulatory effect on the control of blood pressure (BP) in hypertensive animals. The oral administration of RB150 renamed firibastat by WHO, an APA inhibitor prodrug, targeting only the S1 subsite, decreases BP in hypertensive patients from various ethnic origins. To identify new families of potent and selective APA inhibitors, we explored the organization of the APA active site, especially the S2' subsite. By molecular modeling, docking, molecular dynamics simulations and site-directed mutagenesis, we revealed that Arg368 and Arg386, in the S2' subsite of human APA established various types of interactions in major part with the P2' residue but also with the P1' residue of APA inhibitors, required for their nanomolar inhibitory potency. We also demonstrated an important role for Arg368 in APA catalysis, in maintaining the structural integrity of the GAMEN motif, a conserved sequence involved in exopeptidase specificity and optimal positioning of the substrate in monozinc aminopeptidases. This arginine together with the GAMEN motif are key players for the catalytic mechanism of these enzymes.


Assuntos
Glutamil Aminopeptidase/química , Glutamil Aminopeptidase/metabolismo , Sítios de Ligação , Catálise , Dissulfetos/farmacologia , Glutamil Aminopeptidase/antagonistas & inibidores , Glutamil Aminopeptidase/genética , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica/efeitos dos fármacos , Relação Estrutura-Atividade , Especificidade por Substrato , Ácidos Sulfônicos/farmacologia
6.
FASEB J ; 31(2): 687-700, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27815337

RESUMO

Apelin, a (neuro)vasoactive peptide, plays a prominent role in controlling cardiovascular functions and water balance. Because the in vivo apelin half-life is in the minute range, we aimed to identify metabolically stable apelin-17 (K17F) analogs. We generated P92 by classic chemical substitutions and LIT01-196 by original addition of a fluorocarbon chain to the N terminus of K17F. Both analogs were much more stable in plasma (half-life >24 h for LIT01-196) than K17F (4.6 min). Analogs displayed a subnanomolar affinity for the apelin receptor and behaved as full agonists with regard to cAMP production, ERK phosphorylation, and apelin receptor internalization. Ex vivo, these compounds induced vasorelaxation of rat aortas and glomerular arterioles, respectively, precontracted with norepinephrine and angiotensin II, and increased cardiac contractility. In vivo, after intracerebroventricular administration in water-deprived mice, P92 and LIT01-196 were 6 and 160 times, respectively, more efficient at inhibiting systemic vasopressin release than K17F. Administered intravenously (nmol/kg range) in normotensive rats, these analogs potently increased urine output and induced a profound and sustained decrease in arterial blood pressure. In summary, these new compounds, which favor diuresis and improve cardiac contractility while reducing vascular resistances, represent promising candidates for the treatment of heart failure and water retention/hyponatremic disorders.-Gerbier, R., Alvear-Perez, R., Margathe, J.-F., Flahault, A., Couvineau, P., Gao, J., De Mota, N., Dabire, H., Li, B., Ceraudo, E., Hus-Citharel, A., Esteoulle, L., Bisoo, C., Hibert, M., Berdeaux, A., Iturrioz, X., Bonnet, D., Llorens-Cortes, C. Development of original metabolically stable apelin-17 analogs with diuretic and cardiovascular effects.


Assuntos
Fármacos Cardiovasculares/farmacologia , Diuréticos/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Receptores de Apelina , Células CHO , Fármacos Cardiovasculares/química , Cricetinae , Cricetulus , Diuréticos/química , Feminino , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Relação Estrutura-Atividade , Vasoconstrição
7.
J Virol ; 90(3): 1647-56, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608314

RESUMO

UNLABELLED: We have previously reported the construction of a murine leukemia virus-based replication-competent gammaretrovirus (SL3-AP) capable of utilizing the human G protein-coupled receptor APJ (hAPJ) as its entry receptor and its natural receptor, the murine Xpr1 receptor, with equal affinities. The apelin receptor has previously been shown to function as a coreceptor for HIV-1, and thus, adaptation of the viral vector to this receptor is of significant interest. Here, we report the molecular evolution of the SL3-AP envelope protein when the virus is cultured in cells harboring either the Xpr1 or the hAPJ receptor. Interestingly, the dual receptor affinity is maintained even after 10 passages in these cells. At the same time, the chimeric viral envelope protein evolves in a distinct pattern in the apelin cassette when passaged on D17 cells expressing hAPJ in three separate molecular evolution studies. This pattern reflects selection for reduced ligand-receptor interaction and is compatible with a model in which SL3-AP has evolved not to activate hAPJ receptor internalization. IMPORTANCE: Few successful examples of engineered retargeting of a retroviral vector exist. The engineered SL3-AP envelope is capable of utilizing either the murine Xpr1 or the human APJ receptor for entry. In addition, SL3-AP is the first example of an engineered retrovirus retaining its dual tropism after several rounds of passaging on cells expressing only one of its receptors. We demonstrate that the virus evolves toward reduced ligand-receptor affinity, which sheds new light on virus adaptation. We provide indirect evidence that such reduced affinity leads to reduced receptor internalization and propose a novel model in which too rapid receptor internalization may decrease virus entry.


Assuntos
Evolução Molecular Direcionada , Gammaretrovirus/fisiologia , Instabilidade Genômica , Proteínas do Envelope Viral/metabolismo , Tropismo Viral , Internalização do Vírus , Animais , Receptores de Apelina , Linhagem Celular , Gammaretrovirus/genética , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/genética , Receptor do Retrovírus Politrópico e Xenotrópico
8.
Chemistry ; 22(4): 1399-405, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26682530

RESUMO

Herein, we develop a convenient method to facilitate the solution-phase fluorescent labelling of peptides based on the chemoselective acylation of α-hydrazinopeptides. This approach combines the advantages of using commercially available amine-reactive dyes and very mild conditions, which are fully compatible with the chemical sensitivity of the dyes. The usefulness of this approach was demonstrated by the labelling of apelin-13 peptide. Various fluorescent probes were readily synthesized, enabling the rapid optimization of their affinities for the apelin receptor. Thus, the first far-red fluorescent ligand with sub-nanomolar affinity for the apelin receptor was characterized and shown to track the receptor efficiently in living cells by fluorescence confocal microscopy.


Assuntos
Corantes Fluorescentes/química , Hidrazinas/química , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos/síntese química , Receptores Acoplados a Proteínas G/química , Acilação , Hidrazinas/síntese química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ligantes , Peptídeos/química , Receptores Acoplados a Proteínas G/metabolismo
9.
FASEB J ; 29(1): 314-22, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25359495

RESUMO

Apelin is the endogenous ligand of the orphan 7-transmembrane domain GPCR APJ, now named the apelin receptor (ApelinR). Apelin plays a prominent role in body fluid and cardiovascular homeostasis. To better understand the structural organization of the ApelinR, we built 3 homology 3-dimensional (3D) models of the human ApelinR using the validated cholecystokinin receptor-1 3D model or the X-ray structures of the ß2-adrenergic and CXCR4 receptors as templates. Docking of the pyroglutamyl form of apelin 13 (pE13F) into these models revealed the conservation at the bottom of the binding site of a hydrophobic cavity in which the C-terminal Phe of pE13F was embedded. In contrast, at the top of the binding site, depending on the model, different interactions were visualized between acidic residues of the ApelinR and the basic residues of pE13F. Using site-directed mutagenesis, we showed that Asp 92, Glu 172, and Asp 282 of rat ApelinR are key residues in apelin binding by interacting with Lys 8, Arg 2, and Arg 4 of pE13F, respectively. These residues are only seen in the CXCR4-based ApelinR 3D model, further validating this model. These findings bring new insights into the structural organization of the ApelinR and the mode of apelin binding.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Apelina , Receptores de Apelina , Sítios de Ligação/genética , Sequência Conservada , AMP Cíclico/biossíntese , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos/química , Mutagênese Sítio-Dirigida , Conformação Proteica , Ratos , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Homologia Estrutural de Proteína
10.
J Biol Chem ; 289(35): 24599-610, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25012663

RESUMO

Apelin plays a prominent role in body fluid and cardiovascular homeostasis. We previously showed that the C-terminal Phe of apelin 17 (K17F) is crucial for triggering apelin receptor internalization and decreasing blood pressure (BP) but is not required for apelin binding or Gi protein coupling. Based on these findings, we hypothesized that the important role of the C-terminal Phe in BP decrease may be as a Gi-independent but ß-arrestin-dependent signaling pathway that could involve MAPKs. For this purpose, we have used apelin fragments K17F and K16P (K17F with the C-terminal Phe deleted), which exhibit opposite profiles on apelin receptor internalization and BP. Using BRET-based biosensors, we showed that whereas K17F activates Gi and promotes ß-arrestin recruitment to the receptor, K16P had a much reduced ability to promote ß-arrestin recruitment while maintaining its Gi activating property, revealing the biased agonist character of K16P. We further show that both ß-arrestin recruitment and apelin receptor internalization contribute to the K17F-stimulated ERK1/2 activity, whereas the K16P-promoted ERK1/2 activity is entirely Gi-dependent. In addition to providing new insights on the structural basis underlying the functional selectivity of apelin peptides, our study indicates that the ß-arrestin-dependent ERK1/2 activation and not the Gi-dependent signaling may participate in K17F-induced BP decrease.


Assuntos
Arrestinas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fenilalanina/metabolismo , Transdução de Sinais , Animais , Apelina , Sequência de Bases , Primers do DNA , Ativação Enzimática , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Fosforilação , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , beta-Arrestinas
11.
Clin Sci (Lond) ; 127(3): 135-48, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24697296

RESUMO

Hypertension affects one-third of the adult population and is a growing problem due to the increasing incidence of obesity and diabetes. Brain RAS (renin-angiotensin system) hyperactivity has been implicated in the development and maintenance of hypertension in several types of experimental and genetic hypertension animal models. We have identified in the brain RAS that APA (aminopeptidase A) and APN (aminopeptidase N), two membrane-bound zinc metalloproteases, are involved in the metabolism of AngII (angiotensin II) and AngIII (angiotensin III) respectively. The present review summarizes the main findings suggesting that AngIII plays a predominant role in the brain RAS in the control of BP (blood pressure). We first explored the organization of the APA active site by site-directed mutagenesis and molecular modelling. The development and the use in vivo of specific and selective APA and APN inhibitors EC33 and PC18 respectively, has allowed the demonstration that brain AngIII generated by APA is one of the main effector peptides of the brain RAS, exerting a tonic stimulatory control over BP in conscious hypertensive rats. This identified brain APA as a potential therapeutic target for the treatment of hypertension, which has led to the development of potent orally active APA inhibitors, such as RB150. RB150 administered orally in hypertensive DOCA (deoxycorticosteroneacetate)-salt rats or SHRs (spontaneously hypertensive rats) crosses the intestinal, hepatic and blood-brain barriers, enters the brain, generates two active molecules of EC33 which inhibit brain APA activity, block the formation of brain AngIII and normalize BP for several hours. The decrease in BP involves two different mechanisms: a decrease in vasopressin release into the bloodstream, which in turn increases diuresis resulting in a blood volume reduction that participates in the decrease in BP and/or a decrease in sympathetic tone, decreasing vascular resistance. RB150 constitutes the prototype of a new class of centrally acting antihypertensive agents and is currently being evaluated in a Phase Ib clinical trial.


Assuntos
Dissulfetos/uso terapêutico , Glutamil Aminopeptidase/antagonistas & inibidores , Hipertensão/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Ácidos Sulfônicos/uso terapêutico , Angiotensina III/metabolismo , Angiotensina III/fisiologia , Animais , Sítios de Ligação , Pressão Sanguínea , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Ensaios Clínicos Fase I como Assunto , Desenho de Fármacos , Glutamil Aminopeptidase/química , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Inibidores de Proteases/farmacocinética , Ratos
12.
Hum Mol Genet ; 20(2): 301-11, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21036942

RESUMO

Renal tubular dysgenesis (RTD) is a recessive autosomal disease characterized by persistent fetal anuria and perinatal death. During the systematic screening of mutations of the different genes of the renin-angiotensin system associated with RTD, two missense mutations in the renin gene were previously identified, the first affects one of the two catalytic aspartates (D38N) of renin, and the second, S69Y, is located upstream of the 'flap', a mobile ß-hairpin structure which covers the substrate-binding site of renin. Here we report a novel renin mutation leading to the duplication of the tyrosine residue Y15dup, homologous to Y9 in some other aspartyl proteases, which seems to play a crucial role along the activation pathway. The biochemical and cellular mechanisms underlying renin inactivation were investigated. We expressed prorenin constructs harboring the identified point mutations in two established cell lines, able (AtT-20 cells) or unable (CHO cells) to process prorenin to renin and we evaluated the cellular localization of renin mutants and their functional properties. All three mutants were misfolded to different levels, were enzymatically inactive and exhibited abnormal intracellular trafficking. We suggest a misfolding of Y15dup renin, a partial misfolding of D38N prorenin and a misfolding of S69Y prorenin leading to complete absence of secretion. The structural consequences of the renin mutations were estimated by molecular modeling, which suggested some important structural alterations. This is the first characterization of the mechanisms underlying loss of renin function in RTD.


Assuntos
Mutação Puntual , Transporte Proteico , Renina/genética , Renina/metabolismo , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/patologia , Sequência de Aminoácidos , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Feminino , Humanos , Túbulos Renais Proximais/anormalidades , Túbulos Renais Proximais/patologia , Modelos Moleculares , Dados de Sequência Molecular , Gravidez , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Renina/análise , Renina/química , Alinhamento de Sequência
13.
J Biol Chem ; 285(42): 32627-37, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20675385

RESUMO

Apelin is the endogenous ligand of the orphan seven-transmembrane domain (TM) G protein-coupled receptor APJ. Apelin is involved in the regulation of body fluid homeostasis and cardiovascular functions. We previously showed the importance of the C-terminal Phe of apelin 17 (K17F) in the hypotensive activity of this peptide. Here, we show either by deleting the Phe residue (K16P) or by substituting it by an Ala (K17A), that it plays a crucial role in apelin receptor internalization but not in apelin binding or in Gα(i)-protein coupling. Then we built a homology three-dimensional model of the human apelin receptor using the cholecystokinin receptor-1 model as a template, and we subsequently docked K17F into the binding site. We visualized a hydrophobic cavity at the bottom of the binding pocket in which the C-terminal Phe of K17F was embedded by Trp(152) in TMIV and Trp(259) and Phe(255) in TMVI. Using molecular modeling and site-directed mutagenesis studies, we further showed that Phe(255) and Trp(259) are key residues in triggering receptor internalization without playing a role in apelin binding or in Gα(i)-protein coupling. These findings bring new insights into apelin receptor activation and show that Phe(255) and Trp(259), by interacting with the C-terminal Phe of the pyroglutamyl form of apelin 13 (pE13F) or K17F, are crucial for apelin receptor internalization.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fenilalanina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Triptofano/metabolismo , Sequência de Aminoácidos , Animais , Apelina , Receptores de Apelina , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Ensaio Radioligante , Ratos , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
14.
FASEB J ; 24(5): 1506-17, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20040517

RESUMO

Apelin plays a prominent role in body fluid and cardiovascular homeostasis. To explore further upstream the role played by this peptide, nonpeptidic agonists and antagonists of the apelin receptor are required. To identify such compounds that do not exist to date, we used an original fluorescence resonance energy transfer-based assay to screen a G-protein-coupled receptor-focused library of fluorescent compounds on the human EGFP-tagged apelin receptor. This led to isolated E339-3D6 that displayed a 90 nM affinity and behaved as a partial agonist with regard to cAMP production and as a full agonist with regard to apelin receptor internalization. Finally, E339-3D6 induced vasorelaxation of rat aorta precontracted with noradrenaline and potently inhibited systemic vasopressin release in water-deprived mice when intracerebroventricularly injected. This compound represents the first nonpeptidic agonist of the apelin receptor, the optimization of which will allow development of a new generation of vasodilator and aquaretic agents.


Assuntos
Dipeptídeos/farmacologia , Fluoresceínas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Animais , Aorta/efeitos dos fármacos , Receptores de Apelina , Colforsina/farmacologia , AMP Cíclico/metabolismo , Dipeptídeos/química , Dipeptídeos/isolamento & purificação , Avaliação Pré-Clínica de Medicamentos/métodos , Fluoresceínas/química , Fluoresceínas/isolamento & purificação , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Masculino , Camundongos , Ratos , Ratos Endogâmicos WKY , Vasodilatação , Vasopressinas/metabolismo
15.
Nat Commun ; 12(1): 305, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436646

RESUMO

Apelin and arginine-vasopressin (AVP) are conversely regulated by osmotic stimuli. We therefore hypothesized that activating the apelin receptor (apelin-R) with LIT01-196, a metabolically stable apelin-17 analog, may be beneficial for treating the Syndrome of Inappropriate Antidiuresis, in which AVP hypersecretion leads to hyponatremia. We show that LIT01-196, which behaves as a potent full agonist for the apelin-R, has an in vivo half-life of 156 minutes in the bloodstream after subcutaneous administration in control rats. In collecting ducts, LIT01-196 decreases dDAVP-induced cAMP production and apical cell surface expression of phosphorylated aquaporin 2 via AVP type 2 receptors, leading to an increase in aqueous diuresis. In a rat experimental model of AVP-induced hyponatremia, LIT01-196 subcutaneously administered blocks the antidiuretic effect of AVP and the AVP-induced increase in urinary osmolality and induces a progressive improvement of hyponatremia. Our data suggest that apelin-R activation constitutes an original approach for hyponatremia treatment.


Assuntos
Apelina/análogos & derivados , Apelina/metabolismo , Arginina Vasopressina/efeitos adversos , Diurese , Hiponatremia/patologia , Hiponatremia/fisiopatologia , Sequência de Aminoácidos , Animais , Apelina/administração & dosagem , Apelina/sangue , Receptores de Apelina/metabolismo , Arginina Vasopressina/sangue , Glicemia/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Linhagem Celular , Colforsina/farmacologia , AMP Cíclico/biossíntese , Desamino Arginina Vasopressina/farmacologia , Modelos Animais de Doenças , Diurese/efeitos dos fármacos , Eletrólitos/sangue , Meia-Vida , Hiponatremia/sangue , Hiponatremia/urina , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/fisiopatologia , Masculino , Camundongos , Modelos Biológicos , Contração Miocárdica/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Tolvaptan/farmacologia
16.
RSC Med Chem ; 12(8): 1402-1413, 2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34458742

RESUMO

High blood pressure and consequential cardiovascular diseases are among the top causes of death worldwide. The apelinergic (APJ) system has emerged as a promising target for the treatment of cardiovascular issues, especially prevention of ischemia reperfusion (IR) injury after a heart attack or stroke. However, rapid degradation of the endogenous apelin peptides in vivo limits their use as therapeutic agents. Here, we study the effects of simple homologue substitutions, i.e. incorporation of non-canonical amino acids l-cyclohexylalanine (l-Cha) and l-homoarginine (l-hArg), on the proteolytic stability of pyr-1-apelin-13 and apelin-17 analogues. The modified 13-mers display up to 40 times longer plasma half-life than native apelin-13 and in preliminary in vivo assay show moderate blood pressure-lowering effects. The corresponding apelin-17 analogues show pronounced blood pressure-lowering effects and up to a 340-fold increase in plasma half-life compared to the native apelin-17 isoforms, suggesting their potential use in the design of metabolically stable apelin analogues to prevent IR injury.

17.
J Med Chem ; 63(20): 12073-12082, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33001648

RESUMO

Apelin is an important contributor to the renin-angiotensin axis, regulating cardiovascular, metabolic, and neurological functions. Apelin-17 has especially potent cardio-physiological effects but is rapidly degraded in human blood (t0.5 ∼ 4 min). Angiotensin-converting enzyme 2 (ACE-2), neprilysin (NEP), and plasma kallikrein (KLKB1) cleave and inactivate it, with the latter cutting within the arginine-arginine site. Here, we show that analogues with an N-terminal polyethylene glycol (PEG) extension as well as peptide bond isosteres resist KLKB1 cleavage but that only the PEG-extended analogues significantly improve physiologically activity. The PEGylated analogues feature comparatively high log D7.4 values and high plasma protein binding, adding to their stability. An alanine scan of apelin-17 reveals that the integrity and conformational flexibility of the KFRR motif are necessary for cardio-physiological activity. An optimized Cbz-PEG6 analogue is presented that is stable in blood (t0.5 ∼ 18 h), has significant blood-pressure lowering effect, and shows fast recovery of heart function in Langendorff assay.


Assuntos
Apelina/química , Polietilenoglicóis/química , Substâncias Protetoras/química , Apelina/análogos & derivados , Humanos , Conformação Molecular , Estereoisomerismo
18.
Biochem J ; 416(1): 37-46, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18598240

RESUMO

APA (aminopeptidase A; EC 3.4.11.7) is a membrane-bound zinc metallopeptidase, also activated by Ca(2+), involved in the formation of brain angiotensin III, which exerts a tonic stimulatory action on the central control of blood pressure in hypertensive animals. In the present study, in the three-dimensional model of the ectodomain of mouse APA, we docked the specific APA inhibitor glutamate phosphonate, in the presence of Ca(2+). The model showed the presence of one Ca(2+) atom in an hydrophilic pocket corresponding to the S1 subsite in which the lateral chain of the inhibitor is pointing. In this pocket, the Ca(2+) atom was hexaco-ordinated with the acidic side chains of Asp(213) and Asp(218), the carbonyl group of Glu(215) and three water molecules, one of them being engaged in a hydrogen bond with the negatively charged carboxylate side chain of the inhibitor. Mutagenic replacement of Asp(213) and Asp(218) with a conservative residue maintained the ability of mutated APAs to be activated by Ca(2+). However, the replacement by a non-conservative residue abolished this property, demonstrating the crucial role of these residues in Ca(2+) binding. We also showed the involvement of these residues in the strict specificity of APA in the presence of Ca(2+) for N-terminal acidic residues from substrates or inhibitors, since mutagenic replacement of Asp(213) and Asp(218) induced a decrease of the inhibitory potencies of inhibitors homologous with acidic residues. Finally, this led to the rational design of a new potent APA inhibitor, NI926 (K(i)=70 nM), which allowed us to precisely localize Asp(213) at the entrance and Asp(218) at the bottom of the S1 subsite. Taken together, these data provide new insight into the organization and functional role of the APA S1 subsite and will allow the design of pharmacophore of the inhibitor, helpful for the development of a new generation of APA inhibitors as central-acting antihypertensive agents.


Assuntos
Ácido Aspártico/química , Cálcio/metabolismo , Glutamil Aminopeptidase/química , Sequência de Aminoácidos , Aminoácidos/farmacologia , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Sítios de Ligação , Células CHO , Cálcio/farmacologia , Cricetinae , Cricetulus , Inibidores Enzimáticos/farmacologia , Glutamatos/farmacologia , Glutamil Aminopeptidase/antagonistas & inibidores , Glutamil Aminopeptidase/genética , Glutamil Aminopeptidase/metabolismo , Cinética , Camundongos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Organofosfonatos/farmacologia , Especificidade por Substrato , Compostos de Sulfidrila/farmacologia
19.
Mol Biol Cell ; 17(12): 5141-52, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16987961

RESUMO

Phosphoprotein enriched in astrocytes-15 kDa (PEA-15), a phosphoprotein enriched in astrocytes, inhibits both apoptosis and proliferation in normal and cancerous cells. Here, analysis of PEA-15 expression in glioblastoma organotypic cultures revealed low levels of PEA-15 in tumor cells migrating away from the explants, regardless of the expression levels in the originating explants. Because glioblastomas are highly invasive primary brain tumors that can originate from astrocytes, we explored the involvement of PEA-15 in the control of astrocyte migration. PEA-15-/- astrocytes presented an enhanced motility in vitro compared with their wild-type counterparts. Accordingly, NIH-3T3 cells transfected by green fluorescent protein-PEA-15 displayed a reduced migration. Reexpression of PEA-15 restored PEA-15-/- astrocyte motility to wild-type levels. Pharmacological manipulations excluded a participation of extracellular signal-regulated kinase/mitogen-activated protein kinase, phosphatidylinositol 3-kinase/Akt, and calcium/calmodulin-dependent protein kinase II in this effect of PEA-15. In contrast, treatment by bisindolylmaleimide, Gö6976, and rottlerin, and chronic application of phorbol 12-myristate 13-acetate and/or bryostatin-1 indicated that PKC delta mediated PEA-15 inhibition of astrocyte migration. PEA-15-/- astrocytes constitutively expressed a 40-kDa form of PKC delta that was down-regulated upon PEA-15 reexpression. Together, these data reveal a new function for PEA-15 in the inhibitory control of astrocyte motility through a PKC delta-dependent pathway involving the constitutive expression of a catalytic fragment of PKC delta.


Assuntos
Astrócitos/citologia , Movimento Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinase C-delta/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Proteínas Reguladoras de Apoptose , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Peso Molecular , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Cicatrização/fisiologia
20.
Ann N Y Acad Sci ; 1455(1): 12-33, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31236974

RESUMO

The apelinergic pathway has been generating increasing interest in the past few years for its potential as a therapeutic target in several conditions associated with the cardiovascular and metabolic systems. Indeed, preclinical and, more recently, clinical evidence both point to this G protein-coupled receptor as a target of interest in the treatment of not only cardiovascular disorders such as heart failure, pulmonary arterial hypertension, atherosclerosis, or septic shock, but also of additional conditions such as water retention/hyponatremic disorders, type 2 diabetes, and preeclampsia. While it is a peculiar system with its two classes of endogenous ligand, the apelins and Elabela, its intricacies are a matter of continuing investigation to finely pinpoint its potential and how it enables crosstalk between the vasculature and organ systems of interest. In this perspective article, we first review the current knowledge on the role of the apelinergic pathway in the above systems, as well as the associated therapeutic indications and existing pharmacological tools. We also offer a perspective on the challenges and potential ahead to advance the apelinergic system as a target for therapeutic intervention in several key areas.


Assuntos
Apelina/metabolismo , Doenças Cardiovasculares/metabolismo , Doenças Metabólicas/metabolismo , Animais , Humanos , Proteólise , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA