Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 277
Filter
1.
Mar Drugs ; 20(5)2022 May 17.
Article in English | MEDLINE | ID: mdl-35621979

ABSTRACT

Discovering new drug candidates with high efficacy and few side effects is a major challenge in new drug development. The two evolutionarily related peptides oxytocin (OXT) and arginine vasopressin (AVP) are known to be associated with a variety of physiological and psychological processes via the association of OXT with three types of AVP receptors. Over decades, many synthetic analogs of these peptides have been designed and tested for therapeutic applications; however, only a few studies of their natural analogs have been performed. In this study, we investigated the bioactivity and usefulness of two natural OXT/AVP analogs that originate from the marine invertebrate Octopus vulgaris, named octopressin (OTP) and cephalotocin (CPT). By measuring the intracellular Ca2+ or cyclic AMP increase in each OXT/AVP receptor subtype-overexpressing cell, we found that CPT, but not OTP, acts as a selective agonist of human AVP type 1b and 2 receptors. This behavior is reminiscent of desmopressin, the most widely prescribed antidiuretic drug in the world. Similar to the case for desmopressin, a single intravenous tail injection of CPT into Sprague-Dawley rats reduced urine output and increased urinary osmolality. In conclusion, we suggest that CPT has a significant antidiuretic effect and that CPT might be beneficial for treating urological conditions such as nocturia, enuresis, and diabetes insipidus.


Subject(s)
Antidiuretic Agents , Octopodiformes , Oxytocin , Animals , Antidiuretic Agents/pharmacology , Arginine Vasopressin/analogs & derivatives , Deamino Arginine Vasopressin/pharmacology , Felypressin/pharmacology , Octopodiformes/metabolism , Oxytocin/analogs & derivatives , Oxytocin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism
2.
Sci Rep ; 11(1): 15813, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34349143

ABSTRACT

During the development of analgesic tolerance to morphine, the V1b vasopressin receptor has been proposed to bind to ß-arrestin 2 and the µ-opioid receptor to enable their interaction. However, direct evidence of such a high-order complex is lacking. Using bioluminescent resonance energy transfer between a split Nanoluciferase and the Venus fluorescent protein, the NanoBit-NanoBRET system, we found that ß-arrestin 2 closely located near the heteromer µ-V1b receptor in the absence of an agonist and moved closer to the receptor carboxyl-termini upon agonist stimulation. An additive effect of the two agonists for opioid and vasopressin receptors was detected on the NanoBRET between the µ-V1b heteromer and ß-arrestin 2. To increase the agonist response of NanoBRET, the ratio of the donor luminophore to the acceptor fluorophore was decreased to the detection limit of luminescence. In the first phase of access, ß-arrestin 2 was likely to bind to the unstimulated V1b receptor in both its phosphorylated and unphosphorylated forms. In contrast, the second-phase access of ß-arrestin 2 was agonist dependent, indicating a possible pharmacological intervention strategy. Therefore, our efficient method should be useful for evaluating chemicals that directly target the vasopressin binding site in the µ-V1b heteromer to reduce the second-phase access of ß-arrestin 2 and thereby to alleviate tolerance to morphine analgesia.


Subject(s)
Analgesics, Opioid/pharmacology , Morphine/pharmacology , Receptors, Vasopressin/metabolism , beta-Arrestin 2/metabolism , Binding Sites , Drug Tolerance , HEK293 Cells , Humans , Phosphorylation , Receptors, Vasopressin/agonists , beta-Arrestin 2/agonists
3.
Neuropharmacology ; 190: 108565, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33891950

ABSTRACT

Arginine vasopressin (AVP) is a nonapeptide that serves as a neuromodulator in the brain and a hormone in the periphery that regulates water homeostasis and vasoconstriction. The subiculum is the major output region of the hippocampus and an integral component in the networks that processes sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information. Whereas the subiculum expresses high densities of AVP-binding sites and AVP has been shown to increase the synaptic excitability of subicular pyramidal neurons, the underlying cellular and molecular mechanisms have not been determined. We found that activation of V1a receptors increased the excitability of subicular pyramidal neurons via activation of TRPV1 channels and depression of the GIRK channels. V1a receptor-induced excitation of subicular pyramidal neurons required the function of phospholipase Cß, but was independent of intracellular Ca2+ release. Protein kinase C was responsible for AVP-mediated depression of GIRK channels, whereas degradation of phosphatidylinositol 4,5-bisphosphate was involved in V1a receptor-elicited activation of TRPV1 channels. Our results may provide one of the cellular and molecular mechanisms to explain the physiological functions of AVP in the brain.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Receptors, Vasopressin/metabolism , TRPV Cation Channels/metabolism , Action Potentials , Animals , Arginine Vasopressin/pharmacology , Calcium/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Membrane Potentials , Mice , Mice, Knockout , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/metabolism , Pyramidal Cells/drug effects , Receptors, Vasopressin/agonists , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Vasoconstrictor Agents/pharmacology
4.
Med Oncol ; 38(4): 38, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33721131

ABSTRACT

Non-visual arrestins (ß-arrestins) are endocytic proteins that mediate agonist-activated GPCRs internalization and signaling pathways in an independent manner. The involvement of ß-arrestins in cancer invasion and metastasis is increasingly reported. So, it is hypothesized that inhibition of ß-arrestins may diminish the survival chances of cancer cells. This study aimed to evaluate the in vitro impact of inhibiting ß-arrestins on the autophagic and/or apoptotic responsiveness of breast cancer cells. We used Barbadin to selectively inhibit ß-Arr/AP2 interaction in AVP-stimulated V2R receptor of triple-negative breast cancer cells (MDA MB-231). Autophagy was assessed by the microtubule-associated protein 1 light chain 3-II (LC3II), apoptosis was measured by Annexin-V/PI staining and cell cycle distribution was investigated based upon the DNA content using flow cytometry. Barbadin reduced cell viability to 69.1% and increased the autophagy marker LC3II and its autophagic effect disappeared in cells transiently starved in Earle's balanced salt solution (EBSS). Also, Barbadin mildly enhanced the expression of P62 mRNA and arrested 63.7% of cells in G0/G1 phase. In parallel, the drug-induced apoptosis in 29.9% of cells (by AV/PI) and 27.8% of cells were trapped in sub-G1 phase. The apoptotic effect of Barbadin was enhanced when autophagy was inhibited by the PI3K inhibitor (Wortmannin). Conclusively, the data demonstrate the dual autophagic and apoptotic effects of ß-ßArr/AP2 inhibition in triple-negative breast cancer cells. These observations nominate ß-Arrs as selective targets in breast cancer treatment.


Subject(s)
Apoptosis , Autophagy , G1 Phase Cell Cycle Checkpoints , Receptors, Vasopressin/metabolism , beta-Arrestins/antagonists & inhibitors , Adaptor Protein Complex 2/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Phosphatidylinositol 3-Kinases/metabolism , Pyrimidines/pharmacology , Receptors, Vasopressin/agonists , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , beta-Arrestins/genetics , beta-Arrestins/metabolism
5.
Pharmacol Res ; 163: 105272, 2021 01.
Article in English | MEDLINE | ID: mdl-33160069

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) sepsis is a severe condition associated with vascular leakage and poor prognosis. The hemodynamic management of sepsis targets hypotension, but there is no specific treatment available for vascular leakage. Arginine vasopressin (AVP) has been used in sepsis to promote vasoconstriction by activating AVP receptor 1 (V1R). However, recent evidence suggests that increased fluid retention may be associated with the AVP receptor 2 (V2R) activation worsening the outcome of sepsis. Hence, we hypothesized that the inhibition of V2R activation ameliorates the severity of microvascular hyperpermeability during sepsis. The hypothesis was tested using a well-characterized and clinically relevant ovine model of MRSA pneumonia/sepsis and in vitro assays of human lung microvascular endothelial cells (HMVECs). in vivo experiments demonstrated that the treatment of septic sheep with tolvaptan (TLVP), an FDA-approved V2R antagonist, significantly attenuated the sepsis-induced fluid retention and markedly reduced the lung water content. These pathological changes were not affected by the treatment with V2R agonist, desmopressin (DDAVP). Additionally, the incubation of cultured HMVECs with DDAVP, and DDAVP along with MRSA significantly increased the paracellular permeability. Finally, both the DDAVP and MRSA-induced hyperpermeability was significantly attenuated by TLVP. Subsequent protein and gene expression assays determined that the V2R-induced increase in permeability is mediated by phospholipase C beta (PLCß) and the potent permeability factor angiopoietin-2. In conclusion, our results indicate that the activation of the AVP-V2R axis is critical in the pathophysiology of severe microvascular hyperpermeability during Gram-positive sepsis. The use of the antagonist TLVP should be considered as adjuvant treatment for septic patients. The results from this clinically relevant animal study are highly translational to clinical practice.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Pneumonia, Staphylococcal/physiopathology , Receptors, Vasopressin/physiology , Sepsis/physiopathology , Sheep Diseases/physiopathology , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Antidiuretic Agents/therapeutic use , Antidiuretic Hormone Receptor Antagonists/therapeutic use , Capillary Permeability/drug effects , Cells, Cultured , Deamino Arginine Vasopressin/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Hemodynamics/drug effects , Humans , Phospholipase C beta/genetics , Pneumonia, Staphylococcal/drug therapy , Pneumonia, Staphylococcal/veterinary , Receptors, Vasopressin/agonists , Sepsis/drug therapy , Sepsis/veterinary , Sheep , Sheep Diseases/drug therapy , Tolvaptan/therapeutic use
6.
Int J Med Sci ; 17(17): 2751-2762, 2020.
Article in English | MEDLINE | ID: mdl-33162803

ABSTRACT

Background: To date, the effect of vasopressin on organ damages after acute mesenteric ischemia (MI) remains poorly understood. Aims: To investigate the effect of terlipressin, a selective vasopressin V1 receptor agonist, versus norepinephrine on the intestinal and renal injuries after acute MI, and to explore the underlying mechanism of terlipressin. Methods: Acute MI model was produced by clamping the superior mesenteric artery for 1 hour. Immediately after unclamping, terlipressin or norepinephrine was intravenously administered for 2 hours. Meanwhile, in vitro, RAW264.7 cells were treated with lipopolysaccharide or lipopolysaccharide+terlipressin. In addition, wortmannin was used to determine the role of phosphoinositide 3-kinase (PI3K)/ protein kinase B (Akt) pathway in the potential impacts of terlipressin. Results: MI led to severe hypotension, caused notable intestinal and renal impairments and resulted in high mortality, which were markedly improved by terlipressin or norepinephrine. Terlipressin increased mean arterial pressure, decreased intestinal epithelial cell apoptosis, inhibited the generation of M1 macrophage in intestinal and renal tissues, and hindered the release of inflammatory cytokines after MI. Moreover, in cultured macrophages, terlipressin reduced the mRNA level of specific M1 markers and the release of inflammatory cytokines caused by lipopolysaccharide challenge. Wortmannin decreased the expression of PI3K and Akt induced by terlipressin in cells and in tissues, and abolished the above protective effects conferred by terlipressin. Conclusions: Terlipressin or norepinephrine could effectively improve organ damages and mortality after acute MI. Terlipressin elevates blood pressure and inhibits intestinal epithelial apoptosis and macrophage M1 polarization via the PI3K/Akt pathway.


Subject(s)
Acute Kidney Injury/drug therapy , Mesenteric Ischemia/drug therapy , Receptors, Vasopressin/agonists , Reperfusion Injury/drug therapy , Terlipressin/administration & dosage , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Arterial Pressure/drug effects , Disease Models, Animal , Humans , Ileum/blood supply , Ileum/drug effects , Ileum/pathology , Intestinal Mucosa/blood supply , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Kidney/blood supply , Kidney/drug effects , Kidney/pathology , Male , Mesenteric Ischemia/complications , Mesenteric Ischemia/pathology , Norepinephrine/administration & dosage , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Specific Pathogen-Free Organisms , Wortmannin/administration & dosage
7.
J Pharmacol Exp Ther ; 375(2): 286-295, 2020 11.
Article in English | MEDLINE | ID: mdl-32943478

ABSTRACT

Sepsis and septic shock are among the most common causes of death in the intensive care unit; advanced therapeutic approaches are thus urgently needed. Vascular hyperpermeability represents a major manifestation of severe sepsis and is responsible for the ensuing organ dysfunction and failure. Vasopressin V1A receptor (V1AR) agonists have shown promise in the treatment of sepsis, increasing blood pressure, and reducing vascular hyperpermeability. The effects of the selective V1AR-selective agonist selepressin have been investigated in an in vitro model of thrombin-, vascular endothelial growth factor-, angiopoietin 2-, and lipopolysaccharide (LPS)-induced pulmonary microvascular endothelial hyperpermeability. Results suggest that selepressin counteracts the effects of all four endothelial barrier disruptors in a concentration-dependent manner, as reflected in real-time measurements of vascular permeability by means of transendothelial electrical resistance. Further, selepressin protected the barrier integrity against the LPS-mediated corruption of the endothelial monolayer integrity, as captured by VE-cadherin and actin staining. The protective effects of selepressin were abolished by silencing of the vasopressin V1AR, as well as by atosiban, an antagonist of the human V1AR. p53 appears to be involved in mediating these palliative effects, since selepressin strongly induced its expression levels, suppressed the inflammatory RhoA/myosin light chain2 pathway, and triggered the barrier-protective effects of the GTPase Rac1. We conclude that V1AR-selective agonists, such as selepressin, may prove useful in the improvement of endothelial barrier function in the management of severe sepsis. SIGNIFICANCE STATEMENT: A cardinal sign of sepsis, a serious disease with significant mortality and no specific treatment, is pulmonary endothelial barrier dysfunction that leads to pulmonary edema. Here, we present evidence that in cultured human lung microvascular endothelial cells, the synthetic, selective vasopressin V1A receptor agonist selepressin protects against endothelial barrier dysfunction caused by four different edemogenic agents, suggesting a potential role of selepressin in the clinical management of sepsis.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Receptors, Vasopressin/agonists , Cadherins/metabolism , Cardiac Myosins/metabolism , Dose-Response Relationship, Drug , Humans , Myosin Light Chains/metabolism , Receptors, Vasopressin/metabolism , Thrombin/metabolism , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Sci Rep ; 10(1): 7269, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32350300

ABSTRACT

Lipidised analgesic peptide prodrugs self-assemble into peptide nanofibers; with the nanofiber morphology protecting the peptide from plasma degradation and improving therapeutic efficacy. Extending this learning, we hypothesised that a self-assembling lipidized peptide arginine vasopressin (AVP) receptor agonist, that had not been designed as a prodrug, could prove pharmacologically active and control urine production. The only approved AVP receptor agonist, desmopressin is indicated for the treatment of central diabetes insipidus (DI), bedwetting, haemophilia A and von Willebrand disease. Desmopressin is well tolerated by most patients, however adverse effects, such as hyponatraemia and water intoxication necessitate a strict fluid intake, thus motivating the search for alternative DI treatments. Selective V2 receptor agonism is required for anti-DI activity and we hypothesised that our new lipidized peptide (METx) would lead to selective AVP receptor agonism. METx was synthesised and characterised and then tested for activity against the V2, V1a and OT uterine receptors and not tested against the V1b receptor as METx was not expected to cross the blood brain barrier. METx was also tested in vivo in a healthy rat model. METx forms nanofibers and is a partial V2 receptor agonist (determined by measuring MDCK cell line cAMP accumulation), producing 57% of AVP's maximal activity (EC50 = 2.7 nM) and is not a V1a agonist up to a concentration of 1 µM (determined by measuring A7r5 cell line D-myo-inositol-1-phosphate accumulation). METx is a weak OT receptor antagonist, reducing the frequency of OT induced contractions (EC50 = 350 nM) and increasing the OT EC50 from 0.081 nM to 21 nM at a concentration of 600 nM. METx (41 nM) had no effect on spontaneous uterine contractions and METx (100 nM) had no effect on OT induced uterine contractions. Simulated binding studies show that binding avidity to the receptors follows the trend: V2 > OT > V1a. On intravenous injection, a nanoparticle formulation of METx reduced urine production in a healthy rat model in a dose responsive manner, with 40 mg kg-1 METx resulting in no urine production over 4 hours. The lipidized self-assembling peptide - METx - is a selective competitive V2 receptor agonist and an anti-diuretic.


Subject(s)
Antidiuretic Agents , Arginine Vasopressin , Lipopeptides , Receptors, Vasopressin/agonists , Urine , Animals , Antidiuretic Agents/chemical synthesis , Antidiuretic Agents/chemistry , Antidiuretic Agents/pharmacology , Arginine Vasopressin/chemical synthesis , Arginine Vasopressin/chemistry , Arginine Vasopressin/pharmacology , Dogs , Female , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Lipopeptides/pharmacology , Madin Darby Canine Kidney Cells , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Vasopressin/metabolism
9.
Peptides ; 129: 170318, 2020 07.
Article in English | MEDLINE | ID: mdl-32330539

ABSTRACT

Intestinal or mesenteric ischemia generally leads to inflammation and injury, potentially developing hypoxia, causing cell death and tissue necrosis. This in turn can lead to sepsis and shock. Conversely, following shock, the intestinal tract is a main organ to experience ischemic/reperfusion injury. Increased intestinal cell-membrane permeability through mesenteric ischemia provoking bacterial translocation and gut-barrier injury can lead to sepsis and multi-organ failure. Hypotension induced by systemic vasodilation and vascular leak in systemic inflammatory response syndrome and sepsis is countered by immediate fluid resuscitation and vasopressor administration, primarily norepinephrine (NE), with possible arginine vasopressin (AVP) supplementation, an agonist of vasopressin V1A and V2 receptors. Selepressin is a selective V1A-receptor agonist, avoiding potential V2 receptor-associated adverse effects. Selepressin, non-selective AVP, and NE effects on mesenteric blood flow (MBF) and gastric mucosa perfusion (GMP) were compared in control rabbits and a lipopolysaccharide-induced, fluid-resuscitated rabbit endotoxemia model. AVP induced a pronounced decrease in MBF and GMP in non-endotoxemic and endotoxemic rabbits, whereas the reduction after selepressin treatment was significantly less for both indicators in the endotoxemic animals. By contrast, NE increased the MBF and did not affect GMP in both groups. Selepressin and AVP induced a pronounced dose-dependent increase in mesenteric vascular resistance in non-endotoxemic and endotoxemic rabbits, tending to be less in endotoxemic animals, whereas a minor increase in both groups was observed with NE. Therefore, in this safety study, the risk for mesenteric ischemia on selepressin treatment was not inferior to AVP, being less in endotoxemic than in non-endotoxemic animals.


Subject(s)
Arginine Vasopressin/metabolism , Endotoxemia/metabolism , Gastric Mucosa/metabolism , Receptors, Vasopressin/metabolism , Sepsis/metabolism , Animals , Arginine Vasopressin/genetics , Chromatography, High Pressure Liquid , Endotoxemia/genetics , Gastric Mucosa/drug effects , Hypotension/metabolism , Hypotension/pathology , Male , Mesenteric Ischemia/genetics , Mesenteric Ischemia/metabolism , Mesenteric Ischemia/pathology , Rabbits , Receptors, Vasopressin/agonists , Receptors, Vasopressin/genetics , Sepsis/genetics
10.
Vitam Horm ; 113: 259-289, 2020.
Article in English | MEDLINE | ID: mdl-32138951

ABSTRACT

Since its discovery, arginine vasopressin (AVP) was subjected to several modifications with the aim of obtaining novel derivatives with increased potency and selectivity for biomedical use. Desmopressin (dDAVP) is a first generation synthetic analog of AVP with hemostatic and antimetastatic activity. dDAVP acts as a selective agonist of the arginine vasopressin type 2 receptor (AVPR2) present in microvascular endothelium and cancer cells. Considering its selective effects on AVPR2-expressing malignant and vascular tissue, and interesting antitumor profile, dDAVP was used as a lead compound for the development of novel peptide analogs with enhanced anticancer efficacy. After conducting different structure-activity relationship studies to determine key aminoacidic positions for its antitumor activity against AVPR2-expressing malignant cells, dDAVP was rationally modified and a wide panel of synthetic analogs with different sequence and structural modifications was assessed. As a result of this structure-based drug derivatization novel AVP analog [V4Q5]dDAVP (1-deamino-4-valine-5-glutamine-8-d-arginine vasopressin) was selected as the most active candidate and further developed. [V4Q5]dDAVP was evaluated in highly aggressive and metastatic cancer preclinical models deploying enhanced cytostatic, antimetastatic and angiostatic effects in comparison to parental peptide dDAVP. In addition, novel compound demonstrated good tolerability as evaluated in several toxicological studies, and cooperative therapeutic effects after combination with standard-of-care chemotherapy. In summary, due to its ability to inhibit growth and tumor-associated angiogenesis, as well as impairing progression of metastatic disease, AVP analogs such as novel [V4Q5]dDAVP are promising compounds for further development as coadjuvant agents for the management of advance or recurrent cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Deamino Arginine Vasopressin/pharmacology , Neoplasms/drug therapy , Receptors, Vasopressin/agonists , Animals , Disease Models, Animal , Mice
11.
Clin Exp Pharmacol Physiol ; 47(5): 759-764, 2020 05.
Article in English | MEDLINE | ID: mdl-31876005

ABSTRACT

Hyponatraemia, a water-electrolyte disorder diagnosed in patients with subarachnoid haemorrhage (SAH), increases a risk of persistent vasospasm. In majority of cases, hyponatraemia results from inappropriate secretion of vasopressin (AVP). The effect of AVP-associated hyponatraemia on cerebral vasculature is unknown. The present study aimed to elucidate the role of AVP in the response of the middle cerebral artery (MCA) of the rat to hyponatraemia. Isolated, cannulated, and pressurized rat MCAs were perfused/superfused with physiological (Na+  = 144 mmol/L) buffer or low-sodium (Na+  = 121 mmol/L) buffer containing either AVP or angiotensin II (ANG II). ANG II was used to check if the effect of low plasma sodium concentration combined with AVP on the MCA tone is unique to vasopressin. At physiological Na+ concentration, vasopressin (1.4 × 10-11  mol/L) or angiotensin II (10-9  mol/L) resulted in relaxation of the MCA. Substitution of low-sodium for the normal sodium buffer with the same concentration of AVP, resulted in the constriction of the MCA. This effect was absent after removal of the endothelium, administration of vasopressin V1 receptor antagonist or concomitant inhibition of endothelin-1 receptors and synthesis of thromboxane A2. In contrast, no constriction of the MCA in low-sodium buffer was observed when AVP was replaced with ANG II. Our data suggest that presence of vasopressin and low sodium ion concentration results in the change of endothelium phenotype from pro-vasodilatory to pro-vasoconstrictory. This phenomenon may be an overlooked factor contributing to vasospasm in SAH patients with hyponatraemia caused by inappropriate antidiuretic hormone secretion (SIADH).


Subject(s)
Endothelium, Vascular/drug effects , Hyponatremia/physiopathology , Middle Cerebral Artery/drug effects , Sodium/deficiency , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasopressins/pharmacology , Vasospasm, Intracranial/physiopathology , Animals , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hyponatremia/complications , Hyponatremia/metabolism , In Vitro Techniques , Male , Middle Cerebral Artery/metabolism , Middle Cerebral Artery/physiopathology , Rats, Wistar , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism , Thromboxane A2/metabolism , Vasospasm, Intracranial/etiology , Vasospasm, Intracranial/metabolism
12.
Sci Rep ; 9(1): 19295, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848378

ABSTRACT

The neuropeptides oxytocin (OT) and vasopressin (VP) and their G protein-coupled receptors OTR, V1aR, V1bR, and V2R form an important and widely-distributed neuroendocrine signaling system. In mammals, this signaling system regulates water homeostasis, blood pressure, reproduction, as well as social behaviors such as pair bonding, trust and aggression. There exists high demand for ligands with differing pharmacological profiles to study the physiological and pathological functions of the individual receptor subtypes. Here, we present the pharmacological characterization of an arthropod (Metaseiulus occidentalis) OT/VP-like nonapeptide across the human OT/VP receptors. I8-arachnotocin is a full agonist with respect to second messenger signaling at human V2R (EC50 34 nM) and V1bR (EC50 1.2 µM), a partial agonist at OTR (EC50 790 nM), and a competitive antagonist at V1aR [pA2 6.25 (558 nM)]. Intriguingly, I8-arachnotocin activated the Gαs pathway of V2R without recruiting either ß-arrestin-1 or ß-arrestin-2. I8-arachnotocin might thus be a novel pharmacological tool to study the (patho)physiological relevance of ß-arrestin-1 or -2 recruitment to the V2R. These findings furthermore highlight arthropods as a novel, vast and untapped source for the discovery of novel pharmacological probes and potential drug leads targeting neurohormone receptors.


Subject(s)
Arthropods/chemistry , Neuropeptides/agonists , Receptors, Vasopressin/agonists , Vasopressins/agonists , Animals , GTP-Binding Proteins/agonists , Humans , Ligands , Neuropeptides/chemistry , Neuropeptides/pharmacology , Oxytocin/agonists , Oxytocin/chemistry , Oxytocin/pharmacology , Protein Binding/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, Vasopressin/chemistry , Signal Transduction/genetics , Vasopressins/chemistry
13.
Sci Rep ; 9(1): 15480, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664130

ABSTRACT

Oxytocin (OXT) is an important neuromodulator of social behaviors via activation of both oxytocin receptors (OXTR) and vasopressin (AVP) 1a receptors (AVPR1a). Marmosets are neotropical primates with a modified OXT ligand (Pro8-OXT), and this ligand shows significant coevolution with traits including social monogamy and litter size. Pro8-OXT produces more potent and efficacious responses at primate OXTR and stronger behavioral effects than the consensus mammalian OXT ligand (Leu8-OXT). Here, we tested whether OXT/AVP ligands show differential levels of crosstalk at primate AVPR1a. We measured binding affinities and Ca2+ signaling responses of AVP, Pro8-OXT and Leu8-OXT at human, macaque, and marmoset AVPR1a. We found that AVP binds with higher affinity than OXT across AVPR1a, and marmoset AVPR1a show a 10-fold lower OXT binding affinity compared to human and macaque AVPR1a. Both Leu8-OXT and Pro8-OXT produce a less efficacious response than AVP at human AVPR1a and higher efficacious response than AVP at marmoset AVPR1a. These data suggest that OXT might partially antagonize endogenous human AVPR1a signaling and enhance marmoset AVPR1a signaling. These findings aid in further understanding inconsistencies observed following systemic intranasal administration of OXT and provide important insights into taxon-specific differences in nonapeptide ligand/receptor coevolution and behavior.


Subject(s)
Arginine Vasopressin/pharmacology , Leucine/chemistry , Oxytocin/pharmacology , Proline/chemistry , Receptors, Oxytocin/agonists , Receptors, Vasopressin/agonists , Animals , Arginine Vasopressin/chemistry , CHO Cells , Calcium/metabolism , Callithrix , Cricetulus , Humans , Macaca , Oxytocin/chemistry , Receptors, Oxytocin/metabolism , Signal Transduction , Species Specificity
14.
J Med Chem ; 62(10): 4991-5005, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31022340

ABSTRACT

The vasopressin analogue desmopressin (desamino-d-arginine8 vasopressin, dDAVP, 1) is a potent vasopressin 2 (V2) receptor (V2R) agonist approved in many countries for the treatment of diabetes insipidus, primary nocturnal enuresis, nocturia, and coagulation disorders. Since 1 is primarily excreted via the kidneys, an age-related decline in kidney function leads to slower elimination, prolonged antidiuresis, and hyponatremia. In search of novel, potent, selective, and short-acting peptidic V2R agonists, we synthesized a series of C-terminally truncated analogues of [Val4]dDAVP, 2, modified in positions 2, 3, and 7 and/or at the disulfide bridge. The peptides were evaluated for in vitro potency at the human V2 receptor, selectivity versus the related receptors (human vasopressin 1a receptor, human vasopressin 1b receptor, and human oxytocin receptor), and pharmacokinetic profiles in rodents and other higher species. The truncated analogues show excellent potency at the V2R, increased systemic clearance, and shorter half-life in rats. Two compounds 19 (c(Bua-Cpa-Thi-Val-Asn-Cys)-Pro-Agm) and 38 (c(Bua-Cpa-Thi-Val-Asn-Cys)-Pro-d-Arg-NEt2) have been selected for clinical development for nocturia.


Subject(s)
Antidiuretic Agents/chemical synthesis , Antidiuretic Agents/pharmacology , Receptors, Vasopressin/agonists , Animals , Antidiuretic Agents/pharmacokinetics , Deamino Arginine Vasopressin/analogs & derivatives , Deamino Arginine Vasopressin/chemical synthesis , Deamino Arginine Vasopressin/pharmacology , Dose-Response Relationship, Drug , Drug Design , Drug Discovery , Half-Life , Humans , Nocturia/drug therapy , Rats , Receptors, Oxytocin/drug effects , Renal Agents/chemical synthesis , Renal Agents/pharmacology , Structure-Activity Relationship
15.
Crit Care ; 23(1): 91, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30871607

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the effects and safety of vasopressin receptor agonists in patients with septic shock. METHODS: PubMed, EMBASE, and Cochrane library were searched for randomized controlled trials evaluating the effects of vasopressin receptor agonists in septic shock patients. Two reviewers performed literature selection, data extraction, and quality evaluation independently. The primary outcome was mortality. And secondary outcomes included intensive care unit (ICU) length of stay, duration of mechanical ventilation, and incidence of adverse events. In addition, a trial sequential analysis (TSA) was performed. RESULTS: Twenty studies were eligible for meta-analysis. The results showed vasopressin receptor agonists use was associated with reduced mortality (relative risk (RR) 0.92; 95% confidence interval (CI) 0.84 to 0.99; I2 = 0%). Nevertheless, they had no significant effects on ICU length of stay (mean deviation (MD) - 0.08, 95% CI, - 0.68 to 0.52, I2 = 0%) and duration of mechanical ventilation (MD - 0.58, 95% CI - 1.47 to 0.31, I2 = 57%). Additionally, there was no significant difference in total adverse events between two groups (RR 1.28, 95% CI 0.87 to 1.90, I2 = 57%), but vasopressin receptor agonists administration could significantly increase the risk of digital ischemia (RR 4.85, 95% CI 2.81 to 8.39, I2 = 26%). Finally, there was no statistical difference of cardiovascular events (RR 0.91, 95% CI 0.53 to 1.57, I2 = 1%), arrhythmia (0.77, 95% CI 0.48 to 1.23, I2 = 23%), mesenteric ischemia (0.83, 95% CI 0.44 to 1.55, I2 = 0%), diarrhea (2.47, 95% CI 0.77 to 7.96, I2 = 49%), cerebrovascular events (1.36, 95% CI 0.18 to 10.54, I2 = 0%), and hyponatremia (1.47, 95% CI 0.84 to 2.55, I2 = 0%) between two groups. Egger's test showed there was no significant publication bias among studies (P = 0.36). CONCLUSIONS: The use of vasopressin might result in reduced mortality in patients with septic shock. An increased risk of digital ischemia must be taken into account.


Subject(s)
Receptors, Vasopressin/agonists , Shock, Septic/drug therapy , Vasopressins/standards , Hospital Mortality , Humans , Length of Stay , Patient Safety/standards , Shock, Septic/mortality , Vasopressins/therapeutic use
16.
Adv Protein Chem Struct Biol ; 118: 249-272, 2019.
Article in English | MEDLINE | ID: mdl-31928727

ABSTRACT

Biased agonists and pharmacological chaperones have demonstrated their potential to harness G protein-coupled receptor signaling and trafficking, and have collectively opened new possibilities in G protein-coupled receptor drug discovery. Combining pharmacological chaperoning and biased agonism properties into a unique given molecule would be of high therapeutic interest in many human diseases resulting from G protein-coupled receptor mutation and misfolding. This strategy perfectly fits to congenital Nephrogenic Diabetes Insipidus which is a typical conformational disease. In most of the cases, it is associated to inactivating mutations of the renal arginine-vasopressin V2 receptor leading to misfolding and intracellular retention of the receptor, causing the inability of patients to concentrate their urine in response to the antidiuretic hormone. Cell-permeable pharmacological chaperones have been successfully challenged to restore plasma membrane localization of the receptor mutants and to rescue their function. Interestingly, different classes of pharmacological chaperones of the V2 receptor have proven their usefulness and efficacy, such as antagonists, agonists as well as biased agonists. These compounds, particularly small-molecule biased agonists which elicit the V2-induced Gs protein-dependent signaling pathway, but not V2-related arrestin-dependent cell responses, represent a potential therapeutic treatment of this X-linked genetic pathology.


Subject(s)
Diabetes Insipidus, Nephrogenic/drug therapy , Protein Folding , Receptors, Vasopressin/metabolism , Drug Discovery , High-Throughput Screening Assays , Humans , Ligands , Mutation , Receptors, Vasopressin/agonists , Receptors, Vasopressin/genetics
17.
G Ital Nefrol ; 35(6)2018 Dec.
Article in Italian | MEDLINE | ID: mdl-30550035

ABSTRACT

ADH is a hormone secreted by neurohypophysis that plays different roles based on the target organ. At the renal level, this peptide is capable of causing electrolyte-free water absorption, thus playing a key role in the hydro-electrolytic balance. There are pathologies and disorders that jeopardize this balance and, in this field, ADH receptor inhibitors such as Vaptans could play a key role. By inhibiting the activation pathway of vasopressin, they are potentially useful in euvolemic and hypervolemic hypotonic hyponatremia. However, clinical trials in heart failure have not given favourable results on clinical outcomes. Even in SIADH, despite their wide use, there is no agreement by experts on their use. Since vaptans inhibit the cAMP pathway in tubular cells, their use has been proposed to inhibit cystogenesis. A clinical trial has shown favourable effects on ADPKD progression. Because vaptans have been shown to be effective in models of renal cysts disorders other than ADPKD, their use has been proposed in diseases such as nephronophthisis and recessive autosomal polycystic disease. Other possible uses of vaptans could be in kidney transplantation and cardiorenal syndrome. Due to the activity of ADH in coagulation and haemostasis, ADH's activation pathway by Desmopressin Acetate could be a useful strategy to reduce the risk of bleeding in biopsies in patients with haemorrhagic risk.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/therapeutic use , Kidney Diseases/drug therapy , Molecular Targeted Therapy , Neurophysins/agonists , Neurophysins/antagonists & inhibitors , Protein Precursors/agonists , Protein Precursors/antagonists & inhibitors , Receptors, Vasopressin/drug effects , Vasopressins/agonists , Vasopressins/antagonists & inhibitors , Water-Electrolyte Imbalance/drug therapy , Antidiuretic Hormone Receptor Antagonists/pharmacology , Cadaver , Cyclic AMP/physiology , Forecasting , Humans , Hyponatremia/drug therapy , Hyponatremia/physiopathology , Kidney Diseases/physiopathology , Kidney Diseases, Cystic/drug therapy , Kidney Transplantation , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/physiology , Neurophysins/physiology , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/physiopathology , Protein Precursors/physiology , Receptors, Vasopressin/agonists , Second Messenger Systems/drug effects , Tissue Donors , Vasopressins/physiology
18.
J Thorac Cardiovasc Surg ; 156(6): 2098-2107, 2018 12.
Article in English | MEDLINE | ID: mdl-30057184

ABSTRACT

OBJECTIVE: Cardioplegic arrest (CP) and cardiopulmonary bypass (CPB) are associated with vasomotor dysfunction of coronary arterioles in patients with diabetes (DM) undergoing cardiac surgery. We hypothesized that DM may up-regulate vasopressin receptor expression and alter the contractile response of coronary arterioles to vasopressin in the setting of CP/CPB. METHODS: Right atrial tissue samples of patients with DM and without (ND) (n = 8 in each group) undergoing cardiac surgery were harvested before and after CP/CPB. The isolated coronary arterioles (80-150 µm) dissected from the harvested right atrial tissue samples were cannulated and pressurized (40 mm Hg) in a no-flow state. The changes in diameter were measured with video microscopy. The protein expression/localization of vasopressin 1A receptors (V1A) and vasopressin 1B receptors (V1B) in the atrial tissue were measured by immune-blotting and immunohistochemistry. RESULTS: The pre-CP/CPB contractile responses of the coronary arterioles to vasopressin were significantly increased post-CP/CPB in both the ND and DM groups. This effect was more pronounced in the vessels from patients in the DM group than that of vessels from patients in the ND group (P < .05). Vasopressin-induced contractile response of the coronary arterioles was inhibited in the presence of the specific V1A antagonist SR 49059 (10-7 M) in both ND and DM vessels (P < .05). The post-CP/CPB protein levels of V1A were significantly increased compared with pre-CP/CPB values in both the ND and DM groups (P < .05), whereas this increase was greater in DM than that of ND (P < .05). Immunohistochemistry staining further indicates that V1B were mainly expressed in the myocardium but not in vascular smooth muscle. CONCLUSIONS: CP/CPB and DM are both associated with up-regulation in V1 receptor expression/localization in human myocardium. Vasopressin may induce coronary arteriolar constriction via V1A. This alteration may lead to increased coronary arteriolar spasm in patients with DM undergoing CP/CPB and cardiac surgery.


Subject(s)
Arterioles/drug effects , Arterioles/surgery , Coronary Artery Bypass/adverse effects , Coronary Vasospasm/chemically induced , Coronary Vessels/drug effects , Coronary Vessels/surgery , Diabetes Mellitus/physiopathology , Vasoconstriction/drug effects , Vasoconstrictor Agents/toxicity , Vasopressins/toxicity , Aged , Arterioles/metabolism , Arterioles/physiopathology , Cardiopulmonary Bypass/adverse effects , Case-Control Studies , Coronary Vasospasm/metabolism , Coronary Vasospasm/physiopathology , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Diabetes Mellitus/metabolism , Female , Heart Arrest, Induced/adverse effects , Humans , Male , Middle Aged , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism , Signal Transduction/drug effects , Up-Regulation
19.
Neuropsychopharmacology ; 43(10): 2109-2117, 2018 09.
Article in English | MEDLINE | ID: mdl-29875448

ABSTRACT

Social play is a highly rewarding behavior displayed mostly during the juvenile period. We recently showed that vasopressin V1a receptor (V1aR) blockade in the lateral septum (LS) enhances social play in male juvenile rats, but reduces it in females. Here, we determined whether the LS-AVP system modulates dopamine (DA) and/or norepinephrine (NE) neurotransmission in the LS to regulate social play behavior in sex-specific ways. Using microdialysis combined with retrodialysis, we demonstrated that both LS-AVP administration and social play exposure increased extracellular LS-DA release in females, but not in males. Pharmacological blockade of LS-DA receptors reduced social play in both sexes, but required a higher dose in females. This suggests that baseline LS-DA release is sufficient for social play in males, while increased LS-DA release is necessary for social play in females. Administration of a V1aR antagonist into the LS inhibited the social play-induced increase in extracellular LS-DA release in females. Furthermore, co-administration of the DA agonist apomorphine prevented the LS-V1aR blockade-induced decrease in social play in females. This suggests that LS-V1aR blockade reduces social play in females by dampening the rise in LS-DA release. Extracellular LS-NE release was enhanced in response to pharmacological manipulations of the LS-AVP system and to social play in males and/or females, but pharmacological blockade or stimulation of LS-NE receptors did not alter social play in either sex. Overall, we define a mechanism by which the LS-AVP system alters LS-DA neurotransmission differently in males than females resulting in the sex-specific regulation of juvenile social play behavior.


Subject(s)
Dopamine/physiology , Norepinephrine/physiology , Reward , Social Behavior , Vasopressins/metabolism , Animals , Dopamine/metabolism , Female , Male , Microdialysis , Norepinephrine/metabolism , Play and Playthings/psychology , Rats , Rats, Wistar , Receptors, Vasopressin/agonists , Septum of Brain/drug effects , Sex Characteristics , Vasopressins/antagonists & inhibitors , Vasopressins/drug effects
20.
FASEB J ; 32(10): 5520-5531, 2018 10.
Article in English | MEDLINE | ID: mdl-29718707

ABSTRACT

Type-II l-arginine:ureahydrolase, arginase-II (Arg-II), is abundantly expressed in the kidney. The physiologic role played by Arg-II in the kidney remains unknown. Herein, we report that in mice that are deficient in Arg-II (Arg-II-/-), total and membrane-associated aquaporin-2 (AQP2) protein levels were significantly higher compared with wild-type (WT) controls. Water deprivation enhanced Arg-II expression, AQP2 levels, and membrane association in collecting ducts. Effects of water deprivation on AQP2 were stronger in Arg-II-/- mice than in WT mice. Accordingly, a decrease in urine volume and an increase in urine osmolality under water deprivation were more pronounced in Arg-II-/- mice than in WT mice, which correlated with a weaker increase in plasma osmolality in Arg-II-/- mice. There was no difference in vasopressin release under water deprivation conditions between either genotype of mice. Although total AQP2 and phosphorylated AQP2-S256 levels (mediated by PKA) in kidneys under water deprivation conditions were significantly higher in Arg-II-/- mice compared with WT animals, there is no difference in the ratio of AQP2-S256:AQP2. In cultured mouse collecting duct principal mCCDcl1 cells, expression of both Arg-II and AQP2 were enhanced by the vasopressin type 2 receptor agonist, desamino- d-arginine vasopressin (dDAVP). Silencing Arg-II enhanced the expression and membrane association of AQP2 by dDAVP without influencing cAMP levels. In conclusion, in vivo and in vitro experiments demonstrate that Arg-II negatively regulates AQP2 and the urine-concentrating capability in kidneys via a mechanism that is not associated with the modulation of the cAMP pathway.-Huang, J., Montani, J.-P., Verrey, F., Feraille, E., Ming, X.-F., Yang, Z. Arginase-II negatively regulates renal aquaporin-2 and water reabsorption.


Subject(s)
Aquaporin 2/metabolism , Arginase/metabolism , Kidney Tubules, Collecting/metabolism , Water/metabolism , Animals , Aquaporin 2/genetics , Arginase/genetics , Arginine Vasopressin/pharmacology , Cell Line , Cyclic AMP/genetics , Cyclic AMP/metabolism , Kidney Tubules, Collecting/cytology , Mice , Mice, Knockout , Receptors, Vasopressin/agonists , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...