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.
Int J Mol Sci ; 24(2)2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36674841

RESUMO

Acute heart failure (AHF) due to acute myocardial infarction (AMI) is likely to involve cardiogenic shock (CS), with neuro-hormonal activation. A relationship between AHF, CS and vasopressin response is suspected. This study aimed to investigate the implication of vasopressin on hemodynamic parameters and tissue perfusion at the early phase of CS complicating AMI. Experiments were performed on male Wistar rats submitted or not to left coronary artery ligation (AMI and Sham). Six groups were studied Sham and AMI treated or not with either a vasopressin antagonist SR-49059 (Sham-SR, AMI-SR) or agonist terlipressin (Sham-TLP, AMI-TLP). Animals were sacrificed one day after surgery (D1) and after hemodynamic parameters determination. Vascular responses to vasopressin were evaluated, ex vivo, on aorta. AHF was defined by a left ventricular ejection fraction below 40%. CS was defined by AHF plus tissue hypoperfusion evidenced by elevated serum lactate level or low mesenteric oxygen saturation (SmO2) at D1. Mortality rates were 40% in AMI, 0% in AMI-SR and 33% in AMI-TLP. Immediately after surgery, a sharp decrease in SmO2 was observed in all groups. At D1, SmO2 recovered in Sham and in SR-treated animals while it remained low in AMI and further decreased in TLP-treated groups. The incidence of CS among AHF animals was 72% in AMI or AMI-TLP while it was reduced to 25% in AMI-SR. Plasma copeptin level was increased by AMI. Maximal contractile response to vasopressin was decreased in AMI (32%) as in TLP- and SR- treated groups regardless of ligation. Increased vasopressin secretion occurring in the early phase of AMI may be responsible of mesenteric hypoperfusion resulting in tissue hypoxia. Treatment with a vasopressin antagonist enhanced mesenteric perfusion and improve survival. This could be an interesting therapeutic strategy to prevent progression to cardiogenic shock.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Masculino , Ratos , Animais , Choque Cardiogênico/etiologia , Volume Sistólico , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Antagonistas dos Receptores de Hormônios Antidiuréticos/uso terapêutico , Função Ventricular Esquerda , Ratos Wistar , Infarto do Miocárdio/complicações , Infarto do Miocárdio/terapia , Insuficiência Cardíaca/etiologia , Vasopressinas/farmacologia
2.
Am J Physiol Renal Physiol ; 321(3): F305-F321, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34282956

RESUMO

Although vasopressin V1B receptor (V1BR) mRNA has been detected in the kidney, the precise renal localization as well as pharmacological and physiological properties of this receptor remain unknown. Using the selective V1B agonist d[Leu4, Lys8]VP, either fluorescent or radioactive, we showed that V1BR is mainly present in principal cells of the inner medullary collecting duct (IMCD) in the male rat kidney. Protein and mRNA expression of V1BR were very low compared with the V2 receptor (V2R). On the microdissected IMCD, d[Leu4, Lys8]VP had no effect on cAMP production but induced a dose-dependent and saturable intracellular Ca2+ concentration increase mobilization with an EC50 value in the nanomolar range. This effect involved both intracellular Ca2+ mobilization and extracellular Ca2+ influx. The selective V1B antagonist SSR149415 strongly reduced the ability of vasopressin to increase intracellular Ca2+ concentration but also cAMP, suggesting a cooperation between V1BR and V2R in IMCD cells expressing both receptors. This cooperation arises from a cross talk between second messenger cascade involving PKC rather than receptor heterodimerization, as supported by potentiation of arginine vasopressin-stimulated cAMP production in human embryonic kidney-293 cells coexpressing the two receptor isoforms and negative results obtained by bioluminescence resonance energy transfer experiments. In vivo, only acute administration of high doses of V1B agonist triggered significant diuretic effects, in contrast with injection of selective V2 agonist. This study brings new data on the localization and signaling pathways of V1BR in the kidney, highlights a cross talk between V1BR and V2R in the IMCD, and suggests that V1BR may counterbalance in some pathophysiological conditions the antidiuretic effect triggered by V2R activation.NEW & NOTEWORTHY Although V1BR mRNA has been detected in the kidney, the precise renal localization as well as pharmacological and physiological properties of this receptor remain unknown. Using original pharmaceutical tools, this study brings new data on the localization and signaling pathways of V1BR, highlights a cross talk between V1BR and V2 receptor (V2R) in the inner medullary collecting duct, and suggests that V1BR may counterbalance in some pathophysiological conditions the antidiuretic effect triggered by V2R activation.


Assuntos
Receptores de Vasopressinas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Vasopressinas/farmacologia , Animais , Arginina Vasopressina/farmacologia , Masculino , Neurofisinas/efeitos dos fármacos , Precursores de Proteínas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Vasopressinas/metabolismo , Vasopressinas/efeitos dos fármacos
3.
Gen Comp Endocrinol ; 258: 15-32, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29155265

RESUMO

It is now accepted that vasopressin, through V1A/V1B receptors, centrally regulates cognitive functions such as memory, affiliation, stress, fear and depression. However, the respective roles of these receptor isoforms and their contribution to stress-related pathologies remain uncertain. The development of new therapeutic treatments requires a precise knowledge of the distribution of these receptors within the brain, which has been so far hampered by the lack of selective V1B markers. In the present study, we have determined the pharmacological properties of three new potent rat V1B fluorescent ligands and demonstrated that they constitute valuable tools for simultaneous visualization and activation of native V1B receptors in living rat brain tissue. Thus, d[Leu4,Lys-Alexa 647)8]VP (analogue 3), the compound with the best affinity-selectivity/fluorescence ratio for the V1B receptor emerged as the most promising. The rat brain regions most concerned by stress such as hippocampus, olfactory bulbs, cortex and amygdala display the highest V1B fluorescent labelling with analogue 3. In the hippocampus CA2, V1B receptors are located on glutamatergic, not GABAergic neurones, and are absent from astrocytes. Using AVP-EGFP rats, we demonstrate the presence of V1B autoreceptors on AVP-secreting neurones not only in the hypothalamus, but also sparsely in the hippocampus. Finally, using both electrophysiology and visualization of ERK phosphorylation, we show analogue 3-induced activation of the V1B receptor in situ. This will help to analyse expression and functionality of V1B receptors in the brain and contribute to further explore the AVPergic circuitry in normal and pathological conditions.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Corantes Fluorescentes/metabolismo , Receptores de Vasopressinas/metabolismo , Animais , Arginina Vasopressina/metabolismo , Astrócitos/metabolismo , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Hipotálamo/metabolismo , Ligantes , Masculino , Neuroanatomia , Neurônios/metabolismo , Hipófise/citologia , Ratos Sprague-Dawley , Receptores de GABA/metabolismo , Coloração e Rotulagem , Vasopressinas/metabolismo
4.
Am J Physiol Endocrinol Metab ; 312(3): E127-E135, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27998960

RESUMO

Recent epidemiological studies have revealed novel relationships between low water intake or high vasopressin (AVP) and the risk of hyperglycemia and diabetes. AVP V1A and V1B receptors (R) are expressed in the liver and pancreatic islets, respectively. The present study was designed to determine the impact of different levels of circulating AVP on glucose homeostasis in normal Sprague-Dawley rats, as well as the respective roles of V1AR and V1BR. We showed that acute injection of AVP induces a dose-dependent increase in glycemia. Pretreatment with a selective V1AR antagonist, but not a V1BR antagonist, dose-dependently prevented the rise in glycemia. V1BR antagonism did not modify the hyperinsulinemic response, resulting from AVP-induced hyperglycemia, but enhanced the fall in glucagonemia. Acute administration of selective V1AR or V1BR agonists confirmed the involvement of V1AR in the hyperglycemic effect of AVP. In chronic experiments, AVP levels were altered in both directions. Sustained AVP infusion through implantable minipumps induced a time-dependent increase in fasting glycemia, whereas lowering endogenous AVP by increasing water intake had no effect. After 4 wk of AVP infusion, the rise in glycemia amounted to 1.1 mmol/l (P < 0.01) without significant change in insulinemia. This effect was attenuated by cotreatment with a V1AR antagonist. Similar results were observed in lean Zucker rats. These findings demonstrate for the first time a causal link between chronic high AVP and hyperglycemia through V1AR activation and, thus, provide a pathophysiological explanation for the relationship observed in human cohorts between the AVP-hydration axis and the risk of diabetes.


Assuntos
Arginina Vasopressina/farmacologia , Glicemia/efeitos dos fármacos , Glucagon/efeitos dos fármacos , Hiperglicemia/sangue , Receptores de Vasopressinas/efeitos dos fármacos , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Glicemia/metabolismo , Técnicas de Introdução de Genes , Glucagon/sangue , Hiperinsulinismo/sangue , Indóis/farmacologia , Insulina/sangue , Masculino , Imagem Óptica , Pâncreas/metabolismo , Peptídeos Cíclicos/farmacologia , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Receptores de Vasopressinas/agonistas , Receptores de Vasopressinas/metabolismo
5.
Pharmacol Res ; 113(Pt A): 257-264, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27586252

RESUMO

Terlipressin is recommended as a gold standard to treat hepatorenal syndrome complicating liver cirrhosis. It is presented as a specific V1A receptor agonist, beyond its enzymatic conversion into lysine8-Vasopressin (LVP), able to counteract the splanchnic vasodilation. However, the complete pharmacological characterization of this drug with respect to the different vasopressin receptor subtypes is missing. We studied terlipressin intrinsic properties, focusing not only on V1A, but also on other vasopressin receptor subtypes. The experimental studies were conducted on rat and human cellular models. Binding experiments were performed on rat liver membranes and CHO cells transfected with the different human vasopressin receptor subtypes. Agonist status was assessed from inositol phosphate or cyclic AMP assays, and measurement of intracellular calcium variations, performed on cultured vascular smooth muscle cells from rat aorta and human uterine artery and CHO cells. Terlipressin binds to the rat and human V1A receptors with an affinity in the micromolar range, a value 120 fold lower than that of LVP. It induces a rapid and transient intracellular calcium increase, a robust stimulation of phospholipase C but with reduced maximal efficiencies as compared to LVP, indicating a partial V1A agonist property. In addition, terlipressin is also a full agonist of human V2 and V1B receptors, with also a micromomolar affinity. CONCLUSIONS: Terlipressin is a non-selective vasopressin analogue, exhibiting intrinsic agonist properties. Its full V2 receptor agonism may result in renal effects potentially aggravating water retention and hyponatremia of cirrhosis.


Assuntos
Síndrome Hepatorrenal/tratamento farmacológico , Lipressina/análogos & derivados , Pró-Fármacos/farmacologia , Receptores de Vasopressinas/agonistas , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Síndrome Hepatorrenal/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Cirrose Hepática/metabolismo , Lipressina/farmacologia , Masculino , Ratos , Ratos Wistar , Terlipressina , Transfecção/métodos , Vasopressinas/efeitos dos fármacos , Vasopressinas/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(17): E1028-37, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22493236

RESUMO

G protein-coupled receptors (GPCRs) have been shown to activate the mitogen-activated protein kinases, ERK1/2, through both G protein-dependent and -independent mechanisms. Here, we describe a G protein-independent mechanism that unravels an unanticipated role for ß-arrestins. Stimulation of the V2 vasopressin receptor (V2R) in cultured cells or in vivo in rat kidney medullar collecting ducts led to the activation of ERK1/2 through the metalloproteinase-mediated shedding of a factor activating the insulin-like growth factor receptor (IGFR). This process was found to be both Src- and ß-arrestin-dependent. Whereas Src was found to act upstream of the metalloproteinase activation and be required for the release of the IGFR-activating factor, ß-arrestins were found to act downstream of the IGFR transactivation. Unexpectedly, the engagement of ß-arrestins by the IGFR but not by the V2R was needed to promote the vasopressin-stimulated ERK1/2 activation, indicating that a pool of ß-arrestins distinct from those ß-arrestins recruited to the V2R acts downstream of the receptor tyrosine kinase to activate ERK1/2. Such a dual site of action for ß-arrestins helps explain the pleiotropic actions of this scaffolding protein. Given the role that V2R-stimulated ERK1/2 plays in kidney cell proliferation, this transactivation mechanism may have important implications for renal pathophysiology. Still, the role of ß-arrestins downstream of a transactivation event is not limited to the V2R, because we observed a similar involvement for an unrelated GPCR (the platelet-activating factor receptor), indicating that it may be a general mechanism shared among GPCRs.


Assuntos
Arrestinas/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores de Somatomedina/metabolismo , Receptores de Vasopressinas/metabolismo , Ativação Transcricional , Animais , Células Cultivadas , Ativação Enzimática , Medula Renal/citologia , Medula Renal/metabolismo , Ratos , Receptores de Somatomedina/genética , beta-Arrestinas
7.
Biol Psychiatry ; 95(8): 785-799, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38952926

RESUMO

Background: Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. Methods: Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. Results: OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. Conclusions: SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.


Assuntos
Modelos Animais de Doenças , Extinção Psicológica , Medo , Camundongos Knockout , Neurônios , Ocitocina , Síndrome de Prader-Willi , Somatostatina , Vasopressinas , Animais , Ocitocina/farmacologia , Somatostatina/farmacologia , Somatostatina/metabolismo , Medo/efeitos dos fármacos , Medo/fisiologia , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Camundongos , Síndrome de Prader-Willi/fisiopatologia , Síndrome de Prader-Willi/tratamento farmacológico , Vasopressinas/metabolismo , Agressão/efeitos dos fármacos , Agressão/fisiologia , Masculino , Comportamento Social , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/metabolismo , Optogenética , Camundongos Endogâmicos C57BL , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Intrinsicamente Desordenadas
8.
Crit Care ; 15(5): R255, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22026977

RESUMO

INTRODUCTION: Post cardiac surgery vasodilatation (PCSV) is possibly related to a vasopressin deficiency that could relate to chronic stimulation of adeno-hypophysis. To assess vasopressin system activation, a perioperative course of copeptin and vasopressin plasma concentrations were studied in consecutive patients operated on for cardiac surgery. METHODS: Sixty-four consecutive patients scheduled for elective cardiac surgery with cardiopulmonary bypass were studied. Hemodynamic, laboratory and clinical data were recorded before and during cardiopulmonary bypass, and at the eighth postoperative hour (H8). At the same time, blood was withdrawn to determine plasma concentrations of arginine vasopressin (AVP, radioimmunoassay) and copeptin (immunoluminometric assay). PCSV was defined as mean arterial blood pressure < 60 mmHg with cardiac index ≥ 2.2 l/min/m², and was treated with norepinephrine to restore mean blood pressure > 60 mmHg. Patients with PCSV were compared with the other patients (controls). Student's t test, Fisher's exact test, or nonparametric tests (Mann-Whitney, Wilcoxon) were used when appropriate. Correlation between AVP and copeptin was evaluated and receiver-operator characteristic analysis assessed the utility of preoperative copeptin to distinguish between controls and PCSV patients. RESULTS: Patients who experienced PCSV had significantly higher copeptin plasma concentration before cardiopulmonary bypass (P < 0.001) but lower AVP concentrations at H8 (P < 0.01) than controls. PCSV patients had preoperative hyponatremia and decreased left ventricle ejection fraction, and experienced more complex surgery (redo). The area under the receiver-operator characteristic curve of preoperative copeptin concentration was 0.86 ± 0.04 (95% confidence interval = 0.78 to 0.94; P < 0.001). The best predictive value for preoperative copeptin plasma concentration was 9.43 pmol/l with a sensitivity of 90% and a specificity of 77%. CONCLUSIONS: High preoperative copeptin plasma concentration is predictive of PSCV and suggests an activation of the AVP system before surgery that may facilitate depletion of endogenous AVP stores and a relative AVP deficit after surgery.


Assuntos
Ponte Cardiopulmonar/efeitos adversos , Glicopeptídeos/sangue , Período Pré-Operatório , Vasoplegia/etiologia , Idoso , Arginina Vasopressina/sangue , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Vasoplegia/sangue
9.
Anim Cells Syst (Seoul) ; 25(5): 337-346, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745439

RESUMO

In mammals, plasmatic osmolality needs to be stable, and it is highly related to the hydric state of the animals which depends on the activity of the hypothalamic neurohypophysial system and more particularly by vasopressin secretion. Meriones, a desert rodent, can survive even without drinking for more than one month. The mechanism(s) by which they survive under these conditions remains poorly understood. In this study, we examine the water's deprivation consequences on the: (1) anatomy, morphology, and physiology of the hypothalamic supraoptic nucleus, (2) body mass and plasma electrolytes changes in male desert rodents 'Meriones libycus' subjected to water deprivation for 30 days. The effect of water deprivation was evaluated on the structural and cellular organization of the supraoptic nucleus by morphological observations and immunohistochemical approaches, allowing the labeling of AVP but also oxytocin. Our finding demonstrated that upon water deprivation (1) the body weight decreased and reached a plateau after a month of water restriction. (2) The plasmatic osmolality began to decrease and return to values similar to control animals at day 30. (3) The SON, both in hydrated and water-deprived animals, is highly developed.(4) The AVP labeling in the SON increased upon dehydration at variance with OT. These changes observed in body mass and plasma osmolality reveal an important adaptive process of male Meriones in response to prolonged water deprivation. Overall, this animal represents an interesting model for the study of water body homeostasis and the mechanisms underlying the survival of desert rodents to xeric environments.

10.
J Clin Invest ; 131(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33232306

RESUMO

Intellectual and social disabilities are common comorbidities in adolescents and adults with MAGE family member L2 (MAGEL2) gene deficiency characterizing the Prader-Willi and Schaaf-Yang neurodevelopmental syndromes. The cellular and molecular mechanisms underlying the risk for autism in these syndromes are not understood. We asked whether vasopressin functions are altered by MAGEL2 deficiency and whether a treatment with vasopressin could alleviate the disabilities of social behavior. We used Magel2-knockout mice (adult males) combined with optogenetic or pharmacological tools to characterize disease modifications in the vasopressinergic brain system and monitor its impact on neurophysiological and behavioral functions. We found that the activation of vasopressin neurons and projections in the lateral septum were inappropriate for performing a social habituation/discrimination task. Mechanistically, the lack of vasopressin impeded the deactivation of somatostatin neurons in the lateral septum, which predicted social discrimination deficits. Correction of vasopressin septal content by administration or optogenetic stimulation of projecting axons suppressed the activity of somatostatin neurons and ameliorated social behavior. This preclinical study identified vasopressin in the lateral septum as a key factor in the pathophysiology of Magel2-related neurodevelopmental syndromes.


Assuntos
Antígenos de Neoplasias/genética , Transtorno Autístico , Comportamento Animal , Proteínas/genética , Núcleos Septais , Comportamento Social , Vasopressinas , Animais , Antígenos de Neoplasias/metabolismo , Transtorno Autístico/tratamento farmacológico , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Proteínas/metabolismo , Núcleos Septais/metabolismo , Núcleos Septais/fisiopatologia , Vasopressinas/deficiência , Vasopressinas/farmacologia
11.
Endocrinology ; 150(1): 239-50, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18787031

RESUMO

The hypothalamic hormone vasopressin (AVP) has known mitogenic effects on various cell types. This study was designed to determine whether sustained elevated levels of circulating AVP could influence cell proliferation within adult tissues known to express different AVP receptors, including the pituitary, adrenal gland, liver, and kidney. Plasmatic AVP was chronically increased by submitting animals to prolonged hyperosmotic stimulation or implanting them with a AVP-containing osmotic minipump. After several days of either treatment, increased cell proliferation was detected only within the kidney. This kidney cell proliferation was not affected by the administration of selective V1a or V1b receptor antagonists but was either inhibited or mimicked by the administration of a selective V2 receptor antagonist or agonist, respectively. Kidney proliferative cells mostly concerned a subpopulation of differentiated tubular cells known to express the V2 receptors and were associated with the phosphorylation of ERK. These data indicate that in the adult rat, sustained elevated levels of circulating AVP stimulates the proliferation of a subpopulation of kidney tubular cells expressing the V2 receptor, providing the first illustration of a mitogenic effect of AVP via the activation of the V2 receptor subtype.


Assuntos
Arginina Vasopressina/sangue , Túbulos Renais/fisiologia , Receptores de Vasopressinas/fisiologia , Animais , Arginina Vasopressina/farmacologia , Divisão Celular/efeitos dos fármacos , Desamino Arginina Vasopressina/farmacologia , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Masculino , Concentração Osmolar , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio/farmacologia
12.
Crit Care Med ; 37(3): 876-81, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19237891

RESUMO

OBJECTIVE: Terlipressin has been proposed as an alternative treatment to catecholamines to restore blood pressure in septic shock. Terlipressin is considered as a vasopressin prodrug capable of releasing small but sustained amounts of [Lysine] vasopressin (LVP) and to provide prolonged biological effect. However, terlipressin may act as a direct vasopressor beyond its conversion into LVP. We investigated terlipressin direct vasoconstrictive properties and consequences on myocardial perfusion and performance. DESIGN: Experimental studies. SETTINGS: National Research Institute Laboratories. SUBJECTS: Rat aorta and heart, human uterine artery. INTERVENTIONS: Studies of vasoconstriction on isolated vascular rings obtained either from rat aorta or human uterine artery, and of coronary flow, ventricular performance, and heart rhythm on rat hearts using a modified Langendorff heart apparatus. MEASUREMENTS AND MAIN RESULTS: Terlipressin induced a rapid, saturable, and dose-dependent contraction of rat aortas and human uterine arteries. Although the maximal terlipressin-induced vasoconstriction observed on rat arteries was weaker than LVP, or arginine-vasopressin, pharmacologic properties on human arteries, such as full agonism and strong maximal effect (900% of the maximal response obtained with phenylephrine), suggest a high potential of terlipressin to directly vasoconstrict human vessels. Similarly, terlipressin induced a saturable and dose-dependent vasoconstriction of coronary arteries that was reversible and antagonized by selective V1a antagonists. Maximum rates of left ventricle pressure rise (dP/dtmax) and fall (dP/dtmin) decreased both only in proportion to the decrease in coronary flow. CONCLUSIONS: Besides long lasting effect through slow conversion into LVP, terlipressin is a fast acting vasopressor peptide per se that has an impact on coronary circulation and myocardial function.


Assuntos
Circulação Coronária/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/fisiologia , Lipressina/análogos & derivados , Vasoconstritores/farmacologia , Animais , Lipressina/farmacologia , Masculino , Ratos , Ratos Wistar , Terlipressina
13.
Cell Calcium ; 43(1): 95-104, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17555812

RESUMO

Calcium-mobilizing hormones and neurotransmitters are known to affect cell morphology and function including cell differentiation or division. In this study, we examined vasopressin (AVP)-induced morphological changes in a polarized system of rat hepatocytes. Light and electron microscope observations showed that AVP induced microvilli formation and a remodeling of the isolated hepatocyte F-actin submembrane cytoskeleton, these two events being correlated. We showed that these effects were rapid, reversible, observed at nanomolar AVP concentration and mediated by the V(1a) receptor. On polarized multicellular systems of hepatocytes, we observed a rapid reduction of the bile canaliculi lumen at the apical pole and micovilli formation at the basolateral domain with an enlarged F-actin cytoskeleton. Neither activation of protein kinase C nor A via phorbol ester or dibutyryl cAMP induced such rapid morphological changes, at variance with ionomycin, suggesting that AVP-induced intracellular calcium rise plays a crucial role in those effects. By using spectrofluorimetry and cytochemistry, we showed that calcium release from intracellular stores was involved in bile canaliculus contraction, while calcium entry from the extracellular space controlled microvilli formation. Taken together, AVP and calcium-mobilizing agonists differentially regulate physiological hepatocyte plasma membrane events at the basal and the apical domains via topographically specialized calcium-dependent mechanisms.


Assuntos
Arginina Vasopressina/farmacologia , Cálcio/metabolismo , Hepatócitos/ultraestrutura , Citoesqueleto de Actina/ultraestrutura , Actinas/análise , Animais , Canalículos Biliares/efeitos dos fármacos , Canalículos Biliares/ultraestrutura , Polaridade Celular , Células Cultivadas , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Microvilosidades/efeitos dos fármacos , Microvilosidades/ultraestrutura , Ratos , Ratos Wistar
14.
Endocrinology ; 149(9): 4279-88, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18483147

RESUMO

We have previously shown that hyperosmotic stimulation of adult Wistar rats induces local angiogenesis within hypothalamic magnocellular nuclei, in relation to the secretion of vascular endothelial growth factor (VEGF) by the magnocellular neurons. The present study aimed at understanding how osmotic stimulus relates to increased VEGF secretion. We first demonstrate a correlation between increased VEGF secretion and local hypoxia. Osmotic stimulation is known to stimulate the metabolic activity of hypothalamic magnocellular neurons producing arginine vasopressin (AVP) and to increase the secretion of AVP, both by axon terminals into the circulation and by dendrites into the extracellular space. In AVP-deficient Brattleboro rats, the dramatic activation of magnocellular hypothalamic neurons failed to induce hypoxia, VEGF expression, or angiogenesis, suggesting a major role of hypothalamic AVP. A possible involvement of dendritic AVP release is supported by the findings that 1) hypoxia and angiogenesis were not observed in non osmotically stimulated Wistar rats in which circulating AVP was increased by the prolonged infusion of exogenous AVP, 2) contractile arterioles afferent to the magnocellular nuclei were strongly constricted by the perivascular application of AVP via V1a receptors (V1a-R) stimulation, and 3) after the intracerebral or ip administrations of selective V1a-R antagonists to osmotically stimulated rats, hypothalamic hypoxia and angiogenesis were or were not inhibited, respectively. Together, these data strongly suggest that the angiogenesis induced by osmotic stimulation relates to tissue hypoxia resulting from the constriction of local arterioles, via the stimulation of perivascular V1a-R by AVP locally released from dendrites.


Assuntos
Arginina Vasopressina/fisiologia , Dendritos/metabolismo , Hipotálamo/irrigação sanguínea , Hipóxia Encefálica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Vasoconstrição/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos , Arginina Vasopressina/antagonistas & inibidores , Arginina Vasopressina/metabolismo , Arginina Vasopressina/farmacologia , Dendritos/efeitos dos fármacos , Hipotálamo/metabolismo , Hipóxia Encefálica/metabolismo , Injeções Intraventriculares , Masculino , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Osmose , Ratos , Ratos Brattleboro , Ratos Long-Evans , Ratos Wistar , Núcleo Supraóptico/efeitos dos fármacos , Núcleo Supraóptico/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vasoconstrição/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/efeitos dos fármacos
15.
Regul Pept ; 148(1-3): 76-87, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18358546

RESUMO

In this study, we identified the multifunctional protein GC1q-R as a novel vasopressin V(2) receptor (V(2)R) interacting protein. For this purpose, we have developed a proteomic approach combining pull-down assays using a cyclic peptide mimicking the third intracellular loop of V(2)R as a bait and mass spectrometry analyses of proteins isolated from either rat or human kidney tissues or the HEK 293 cell line. Co-immunoprecipitation of GC1q-R with the c-Myc-tagged h-V(2)R expressed in a HEK cell line confirmed the existence of a specific interaction between GC1q-R and the V(2) receptor. Then, construction of a mutant receptor in i3 loop allowed us to identify the i3 loop arginine cluster of the vasopressin V(2) receptor as the interacting determinant for GC1q-R interaction. Using purified receptor as a bait and recombinant (74-282) GC1q-R, we demonstrated a direct and specific interaction between these two proteins via the arginine cluster.


Assuntos
Arginina/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Mitocondriais/metabolismo , Receptores de Vasopressinas/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Arginina/química , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Humanos , Rim/metabolismo , Pessoa de Meia-Idade , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Ratos , Receptores de Vasopressinas/química , Receptores de Vasopressinas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
16.
Mol Endocrinol ; 21(2): 512-23, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17082326

RESUMO

Starting from the 2.8-A resolution x-ray structure of bovine rhodopsin, three-dimensional molecular models of the complexes between arginine vasopressin and two receptor subtypes (V1a, V1b) have been built. Amino acid sequence alignment and docking studies suggest that four key residues (1.35, 2.65, 4.61, and 5.35) fine tune the binding of vasopressin and related peptide agonists to both receptor subtypes. To validate these predictions, a series of single or double mutants were engineered at V1a and V1b receptor subtypes and tested for their binding and functional properties. Two negatively charged amino acids at positions 1.35 and 2.65 are key anchoring residues to the Arg8 residue of arginine vasopressin. Moreover, two amino acids (V(4.61) and P(5.35)) delineating a hydrophobic subsite at the human V1b receptor are responsible for the recognition of V1b selective peptide agonists. Last, one of the latter positions (5.35) is hypothesized to explain the pharmacological species differences between rat and human vasopressin receptors for a V1b peptide agonist. Altogether these refined three-dimensional models of V1a and V1b human receptors should enable the identification of further new selective V1a and V1b agonists as pharmacological but also therapeutic tools.


Assuntos
Arginina Vasopressina/química , Modelos Moleculares , Receptores de Vasopressinas/química , Sequência de Aminoácidos , Animais , Arginina Vasopressina/metabolismo , Sítios de Ligação , Células CHO , Bovinos , Cricetinae , Cricetulus , Humanos , Dados de Sequência Molecular , Mutação , Conformação Proteica , Mapeamento de Interação de Proteínas , Ensaio Radioligante , Ratos , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Homologia de Sequência de Aminoácidos
17.
Endocrinology ; 148(9): 4136-46, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17495006

RESUMO

Recently, we synthesized and characterized the first selective V(1b) vasopressin (VP)/oxytocin receptor agonist, d[Cha(4)]arginine vasopressin. However, this agonist was only selective for the human receptors. We thus decided to design a selective V(1b) agonist for the rodent species. We started from previous observations showing that modifying [deamino(1),Arg(8)]VP in positions 4 and 8 altered the rat VP/oxytocin receptor selectivity. We synthesized a series of 13 [deamino(1),Arg(8)]VP analogs modified in positions 4 and 8. Among them, one seemed very promising, d[Leu(4), Lys(8)]VP. In this paper, we describe its pharmacological and physiological properties. This analog exhibited a nanomolar affinity for the rat, human, and mouse V(1b) VP receptors and a strong V(1b) selectivity for the rat species. On AtT20 cells stably transfected with the rat V(1b) receptor, d[Leu(4), Lys(8)]VP behaved as a full agonist on both phospholipase C and MAPK assays. Additional experiments revealed its ability to induce the internalization of enhanced green fluorescent protein-tagged human and mouse V(1b) receptors as expected for a full agonist. Additional physiological experiments were performed to further confirm the selectivity of this peptide. Its antidiuretic, vasopressor, and in vitro oxytocic activities were weak compared with those of VP. In contrast, used at low doses, its efficiency to stimulate adrenocorticotropin or insulin release from mouse pituitary or perfused rat pancreas, respectively, was similar to that obtained with VP. In conclusion, d[Leu(4), Lys(8)]VP is the first selective agonist available for the rat V(1b) VP receptor. It will allow a better understanding of V(1b) receptor-mediated effects in rodents.


Assuntos
Lipressina/análogos & derivados , Receptores de Ocitocina/agonistas , Receptores de Vasopressinas/agonistas , Adenilil Ciclases/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Feminino , Humanos , Rim/efeitos dos fármacos , Rim/fisiologia , Lactação , Fígado/efeitos dos fármacos , Fígado/fisiologia , Lipressina/síntese química , Lipressina/farmacologia , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/fisiologia , Camundongos , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/fisiologia , Ratos , Ratos Wistar , Receptores de Ocitocina/efeitos dos fármacos , Receptores de Ocitocina/genética , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/efeitos dos fármacos , Transfecção
18.
J Med Chem ; 50(4): 835-47, 2007 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-17300166

RESUMO

The neurohypophyseal peptides arginine vasopressin (AVP) and oxytocin (OT) mediate a wide variety of peripheral and central physiological and behavioral effects by acting on four different G-protein coupled receptors, termed V1a (vascular), V1b (pituitary), V2 (renal), and OT (uterine). We recently reported that d[Cha4]AVP (A), d[Leu4]AVP (B), d[Orn4]AVP (C), and d[Arg4]AVP (D) have high affinity and are selective agonists for the human V1b receptor. However, peptides A-D were subsequently shown to be potent antidiuretic agonists in the rat and are, thus, not selective V1b agonists in the rat. Peptides A-D served as leads for the studies reported here. They were modified at position 8 by Lys, ornithine (Orn), diaminobutyric acid (Dab), and diaminopropionic acid (Dap) to give d[Cha4,Lys8]VP (1), d[Cha4,Orn8]VP (2), d[Cha4,Dab8]VP (3), d[Cha4,Dap8]VP (4), d[Leu4,Lys8]VP (5), d[Leu4,Orn8]VP (6), d[Leu4,Dab8]VP (7), d[Leu4,Dap8]VP (8), d[Orn4,Lys8]VP (9), d[Orn4,Orn8]VP (10), d[Arg4,Lys8]VP (11), d[Arg4,Orn8]VP (12), and d[Arg4,Dab8]VP (13). All peptides were synthesized by the Merrifield solid-phase method. Their binding and functional properties were evaluated in rat AVP V1a, V1b, and V2 receptors and on the rat OT receptor expressed either in native tissues or in stably transfected cells. They were also examined in rat vasopressor, antidiuretic, and in in vitro (no Mg++) oxytocic assays. Functional studies performed on chinese hamster ovary cells expressing the different AVP/OT receptors confirm that d[Cha4,Lys8]VP (1), d[Cha4,Dab8]VP (3), d[Leu4,Lys8]VP (5), and d[Leu4,Dap8]VP (8) are the first selective agonists for the rat V1b receptor. These selective V1b agonists are promising new tools for studies of the role of the V1b receptor in the rat.


Assuntos
Arginina Vasopressina/análogos & derivados , Arginina Vasopressina/síntese química , Oligopeptídeos/síntese química , Peptídeos Cíclicos/síntese química , Receptores de Vasopressinas/agonistas , Adenilil Ciclases/metabolismo , Animais , Antidiuréticos/farmacologia , Arginina Vasopressina/farmacologia , Linhagem Celular , Cricetinae , Cricetulus , Desenho de Fármacos , Técnicas In Vitro , Fosfatos de Inositol/biossíntese , Oligopeptídeos/farmacologia , Ocitócicos/farmacologia , Peptídeos Cíclicos/farmacologia , Isoformas de Proteínas/agonistas , Ensaio Radioligante , Ratos , Receptores de Ocitocina/agonistas , Relação Estrutura-Atividade , Vasoconstritores/farmacologia
19.
Mol Cell Biol ; 36(6): 1019-31, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26787837

RESUMO

Palmitoylation is involved in several neuropsychiatric and movement disorders for which a dysfunctional signaling of the dopamine D3 receptor (Drd3) is hypothesized. Computational modeling of Drd3's homologue, Drd2, has shed some light on the putative role of palmitoylation as a reversible switch for dopaminergic receptor signaling. Drd3 is presumed to be palmitoylated, based on sequence homology with Drd2, but the functional attributes afforded by Drd3 palmitoylation have not been studied. Since these receptors are major targets of antipsychotic and anti-Parkinsonian drugs, a better characterization of Drd3 signaling and posttranslational modifications, like palmitoylation, may improve the prospects for drug development. Using molecular dynamics simulations, we evaluated in silico how Drd3 palmitoylation could elicit significant remodeling of the C-terminal cytoplasmic domain to expose docking sites for signaling proteins. We tested this model in cellulo by using the interaction of Drd3 with the G-alpha interacting protein (GAIP) C terminus 1 (GIPC1) as a template. From a series of biochemical studies, live imaging, and analyses of mutant proteins, we propose that Drd3 palmitoylation acts as a molecular switch for Drd3-biased signaling via a GIPC1-dependent route, which is likely to affect the mode of action of antipsychotic drugs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Palmitatos/metabolismo , Receptores de Dopamina D3/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/genética , Membrana Celular/metabolismo , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Mapas de Interação de Proteínas , Transporte Proteico , Receptores de Dopamina D3/análise , Receptores de Dopamina D3/genética , Transdução de Sinais
20.
J Neurosci ; 24(2): 370-7, 2004 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-14724235

RESUMO

G-protein-coupled receptors (GPCRs) play a major role in cell-cell communication in the CNS. These proteins oscillate between various inactive and active conformations, the latter being stabilized by agonists. Although mutations can lead to constitutive activity, most of these destabilize inactive conformations, and none lock the receptor in an active state. Moreover, GPCRs are known to form dimers, but the role of each protomer in the activation process remains unclear. Here, we show that the heterodimeric GPCR for the main inhibitory neurotransmitter, the GABA(B) receptor, can be locked in its active state by introducing two cysteines expected to form a disulphide bridge to maintain the binding domain of the GABA(B1) subunit in a closed form. This constitutively active receptor cannot be inhibited by antagonists, but its normal functioning, activation by agonists, and inhibition by antagonists can be restored after reduction with dithiothreitol. These data show that the closed state of the binding domain of GABA(B1) is sufficient to turn ON this heterodimeric receptor and illustrate for the first time that a GPCR can be locked in an active conformation.


Assuntos
Receptores de GABA-B/metabolismo , Linhagem Celular , Cisteína/genética , Dimerização , Dissulfetos/química , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Humanos , Modelos Moleculares , Mutação , Estrutura Terciária de Proteína , Receptores de GABA-B/química , Receptores de GABA-B/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa