1
|
Michael OS, Kanthakumar P, Soni H, Rajesh Lenin R, Abhiram Jha K, Gangaraju R, Adebiyi A. Urotensin II system in chronic kidney disease. Curr Res Physiol 2024; 7:100126. [PMID: 38779598 PMCID: PMC11109353 DOI: 10.1016/j.crphys.2024.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/23/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic kidney disease (CKD) is a progressive and long-term condition marked by a gradual decline in kidney function. CKD is prevalent among those with conditions such as diabetes mellitus, hypertension, and glomerulonephritis. Affecting over 10% of the global population, CKD stands as a significant cause of morbidity and mortality. Despite substantial advances in understanding CKD pathophysiology and management, there is still a need to explore novel mechanisms and potential therapeutic targets. Urotensin II (UII), a potent vasoactive peptide, has garnered attention for its possible role in the development and progression of CKD. The UII system consists of endogenous ligands UII and UII-related peptide (URP) and their receptor, UT. URP pathophysiology is understudied, but alterations in tissue expression levels of UII and UT and blood or urinary UII concentrations have been linked to cardiovascular and kidney dysfunctions, including systemic hypertension, chronic heart failure, glomerulonephritis, and diabetes. UII gene polymorphisms are associated with increased risk of diabetes. Pharmacological inhibition or genetic ablation of UT mitigated kidney and cardiovascular disease in rodents, making the UII system a potential target for slowing CKD progression. However, a deeper understanding of the UII system's cellular mechanisms in renal and extrarenal organs is essential for comprehending its role in CKD pathophysiology. This review explores the evolving connections between the UII system and CKD, addressing potential mechanisms, therapeutic implications, controversies, and unexplored concepts.
Collapse
Affiliation(s)
- Olugbenga S. Michael
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Praghalathan Kanthakumar
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hitesh Soni
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Raji Rajesh Lenin
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kumar Abhiram Jha
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Adebowale Adebiyi
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| |
Collapse
|
2
|
Zhang NN, Xu HY, Liu XN, Chen YF, Xia CM, Wu XZ, Lu N. The Inhibitory Role of Hydrogen Sulfide in UII-Induced Cardiovascular Effects and the Underlying Signaling Pathways. Antioxidants (Basel) 2022; 11:2253. [PMID: 36421438 PMCID: PMC9686774 DOI: 10.3390/antiox11112253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 12/08/2023] Open
Abstract
Urotensin II (UII) could increase blood pressure and heart rate via increased central reactive oxygen species (ROS) levels. We reported previously that hydrogen sulfide (H2S) exerts an antihypertensive effect by suppressing ROS production. The aim of the current study is to further examine the effects of endogenous and exogenous H2S on UII-induced cardiovascular effects by using an integrated physiology approach. We also use cell culture and molecular biological techniques to explore the inhibitory role of H2S on UII-induced cardiovascular effects. In this study, we found that cystathionine-β-synthase (CBS), the main H2S synthesizing enzyme in CNS, was expressed in neuronal cells of the rostral ventrolateral medulla (RVLM) area. Cellular distribution of CBS and urotensin II receptor (UT) in SH-SY5Y cells that are confirmed as glutamatergic were identified by immunofluorescent and Western blots assay. In Sprague-Dawley rats, administration of UII into the RVLM resulted in an increase in mean arterial pressure (MAP), heart rate (HR), ROS production, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and phosphorylation of p47phox, extracellular signal-regulated protein kinase (ERK)1/2 and p38MAPK, but not stress-activated protein kinase/Jun N-terminal kinase (SAPK/JNK). These effects of UII were attenuated by application into the RVLM of endogenous (L-cysteine, SAM) or exogenous (NaHS) H2S. These results were confirmed in SH-SY5Y cells. UII-induced cardiovascular effects were also significantly abolished by pretreatment with microinjection of Tempol, Apocynin, SB203580, or PD98059 into the RVLM. Preincubated SH-SY5Y cells with Apocynin before administration of UII followed by Western blots assay showed that ROS is in the upstream of p38MAPK/ERK1/2. Gao activation assay in SH-SY5Y cells suggested that H2S may exert an inhibitory role on UII-induced cardiovascular effects by inhibiting the activity of Gαo. These results suggest that both endogenous and exogenous H2S attenuate UII-induced cardiovascular effects via Gαo-ROS-p38MAPK/ERK1/2 pathway.
Collapse
Affiliation(s)
- Na-Na Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Department of Urology, Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hai-Yan Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiao-Ni Liu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yi-Fan Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chun-Mei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xing-Zhong Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ning Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
3
|
Microinjection of urotensin II into the rostral ventrolateral medulla increases sympathetic vasomotor tone via the GPR14/ERK pathway in rats. Hypertens Res 2020; 43:765-771. [PMID: 32385485 DOI: 10.1038/s41440-020-0460-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/20/2020] [Accepted: 03/12/2020] [Indexed: 01/17/2023]
Abstract
The present study aimed to reveal the effects of urotensin II (UII) on sympathetic vasomotor tone in the rostral ventrolateral medulla (RVLM). UII (0.3, 3, and 30 nmol/L, 50 nL) was microinjected into the RVLM. Blood pressure (BP), heart rate (HR), and renal sympathetic nerve activity (RSNA) were measured to determine the sympathetic vasomotor tone. BP, HR, and RSNA were simultaneously recorded after drugs had been microinjected into the RVLM. Microinjection of UII (0.3, 3, and 30 nmol/L, 50 nL) into the RVLM significantly increased BP, HR, and RSNA. Pretreatment with BIM23127 (300 nmol/L, 50 nL), a potent antagonist of the UII receptor GPR14, abolished the effect of UII. Previous microinjection of PD98059 (25 μmol/L, 50 nL), an inhibitor of ERK, significantly suppressed the effects of UII. Preinjection of an inhibitor of the N-type Ca2+ channel, ω-conotoxin GVIA (50 nmol/L, 50 nL), inhibited the effects of UII. The present study demonstrated that microinjection of UII into the RVLM significantly increased sympathetic vasomotor tone, which was mediated by the GPR14/ERK/N-type Ca2+ channel pathway. UII may become a novel therapeutic target for autonomic nervous system regulation, especially in hypertension.
Collapse
|
4
|
Ettaro R, Markovic T, Daniels D, MacLaren DA, Clark SD. Microinjection of urotensin II into the pedunculopontine tegmentum leads to an increase in the consumption of sweet tastants. Physiol Behav 2020; 215:112775. [PMID: 31843472 DOI: 10.1016/j.physbeh.2019.112775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/02/2019] [Accepted: 12/11/2019] [Indexed: 11/29/2022]
Abstract
The pedunculopontine tegmentum (PPTg) plays a role in processing multiple sensory inputs and innervates brain regions associated with reward-related behaviors. The urotensin II receptor, activated by the urotensin II peptide (UII), is selectively expressed by the cholinergic neurons of the PPTg. Although the exact function of cholinergic neurons of the PPTg is unknown, they are thought to contribute to the perception of reward magnitude or salience detection. We hypothesized that the activation of PPTg cholinergic neurons would alter sensory processing across multiple modalities (ex. taste and hearing). Here we had three aims: first, determine if cholinergic activation is involved in consumption behavior of palatable solutions (sucrose). Second, if so, distinguish the impact of the caloric value by using saccharin, a zero calorie sweetener. Lastly, we tested the UII-mediated effects on perception of acoustic stimuli by measuring acoustic startle reflex (ASR). Male Sprague-Dawley rats were bilaterally cannulated into the PPTg, then placed under food restriction lasting the entire consumption experiment (water ad lib.). Treatment consisted of a microinjection of either 1 μL of aCSF or 1 μL of 10 μM UII into the PPTg, and the rats were immediately given access to either sucrose or saccharin. For the remaining five days, rats were allowed one hour access per day to the same sweet solution without any further treatments. During the saccharin experiment rats were tested in a contact lickometer which recorded each individual lick to give insight into the microstructure of the consumption behavior. ASR testing consisted of a baseline (no treatment), treatment day, and two additional days (no treatment). Immediately following the microinjection of UII, consumption of both saccharin and sucrose increased compared to controls. This significant increase persisted for days after the single administration of UII, but there was no generalized arousal or increase in water consumption between testing sessions. The effects on ASR were not significant. Activating cholinergic PPTg neurons may lead to a miscalculation of the salience of external stimuli, implicating the importance of cholinergic input in modulating a variety of behaviors. The long-lasting effects seen after UII treatment support further research into the role of sensory processing on reward related-behaviors at the level of the PPTg cholinergic neurons.
Collapse
Affiliation(s)
- Robert Ettaro
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| | - Tamara Markovic
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| | - Derek Daniels
- Department of Psychology and the Center for Ingestive Behavior Research, University at Buffalo, Buffalo, NY 14214, United States
| | - Duncan Aa MacLaren
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| | - Stewart D Clark
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, United States
| |
Collapse
|
5
|
Pereira-Castro J, Brás-Silva C, Fontes-Sousa AP. Novel insights into the role of urotensin II in cardiovascular disease. Drug Discov Today 2019; 24:2170-2180. [PMID: 31430542 DOI: 10.1016/j.drudis.2019.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/26/2019] [Accepted: 08/12/2019] [Indexed: 12/16/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide that interacts with a specific receptor called the UT receptor. UII has been implicated in cardiovascular regulation, with promising therapeutic applications based on UT receptor antagonism. The endogenous ligands of the UT receptor: UII and urotensin-related peptide (URP), differentially bind and activate this receptor. Also, the receptor localization is not restricted to the plasma membrane, possibly inducing different physiological responses that could support its inconsistent, but potent, vasoactive activity. These properties could explain the disappointing outcomes in clinical studies, in contrast to the positive preclinical results regarding heart failure, pulmonary hypertension, atherosclerosis and diabetes mellitus. These aspects should be considered in future investigations to a better comprehension of the role of UII as a potential therapeutic target.
Collapse
Affiliation(s)
- João Pereira-Castro
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, UnIC - Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ana Patrícia Fontes-Sousa
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal.
| |
Collapse
|
6
|
Lim K, Sata Y, Jackson KL, Burke SL, Head GA. Acute Effect of Central Administration of Urotensin II on Baroreflex and Blood Pressure in Conscious Normotensive Rabbits. Front Physiol 2017; 8:110. [PMID: 28280470 PMCID: PMC5322237 DOI: 10.3389/fphys.2017.00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/09/2017] [Indexed: 12/03/2022] Open
Abstract
In the present study, we examined the effects of central administration of Urotensin II on blood pressure, heart rate, and baroreceptor heart rate reflexes in conscious normotensive rabbits. Preliminary operations were undertaken to implant a balloon cuff on the inferior vena cava for baroreflex assessments and to implant cannula into the lateral and fourth ventricle. After 2 weeks of recovery cumulative dose response curves to Urotensin II (10, 100 ng, 1, 10, and 100 μg) given into the ventricles, or Ringer's solution as a vehicle were performed on separate days. Injections were given each hour and baroreflex assessments were made 30 min after each administration. Analysis of the dose response curves to Urotensin II compared to vehicle administered into the lateral or fourth ventricle, indicated little change to blood pressure or heart rate. Analysis of the time course to the highest dose over a 30 min period revealed a small (−5 mmHg) depressor response maximal at 10 min when injected into the fourth ventricle but no effect when injected into the lateral ventricle. Baroreflex assessments made at each dose showed that there was no change in baroreflex sensitivity but that an increase in the upper plateau was observed when Urotensin was injected into the lateral ventricle and a tendency for a reduced lower heart rate plateau was observed after fourth ventricle administration. Clonidine administration in the fourth ventricle decreased blood pressure and heart rate, thus confirming catheter patency. In conclusion, our findings suggest that Urotensin II in the forebrain and brainstem may play a role in modulating cardiac sympathetic and vagal baroreflexes but only during large acute changes in blood pressure.
Collapse
Affiliation(s)
- Kyungjoon Lim
- Department of Neuropharmacology, Baker IDI Heart and Diabetes Research InstituteMelbourne, VIC, Australia; Department of Physiology, Monash UniversityClayton, VIC, Australia
| | - Yusuke Sata
- Department of Neuropharmacology, Baker IDI Heart and Diabetes Research InstituteMelbourne, VIC, Australia; Faculty of Medicine, Nursing and Health Science, Monash UniversityClayton, VIC, Australia
| | - Kristy L Jackson
- Department of Neuropharmacology, Baker IDI Heart and Diabetes Research Institute Melbourne, VIC, Australia
| | - Sandra L Burke
- Department of Neuropharmacology, Baker IDI Heart and Diabetes Research Institute Melbourne, VIC, Australia
| | - Geoffrey A Head
- Department of Neuropharmacology, Baker IDI Heart and Diabetes Research InstituteMelbourne, VIC, Australia; Department of Pharmacology, Monash UniversityClayton, VIC, Australia
| |
Collapse
|
7
|
Şatıroğlu Ö, Durakoğlugil ME, Çetin M, Çiçek Y, Erdoğan T, Duman H. The role of urotensin II and atherosclerotic risk factors in patients with slow coronary flow. Interv Med Appl Sci 2016; 8:158-163. [PMID: 28180005 PMCID: PMC5283774 DOI: 10.1556/1646.8.2016.4.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 08/07/2016] [Accepted: 09/20/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Slow coronary flow (SCF) is an angiographic finding characterized with delayed opacification of epicardial coronary arteries without obstructive coronary disease. Urotensin II (UII) is an important vascular peptide, which has an important role in hypertension, coronary artery disease, and vascular remodeling in addition to potent vasoconstrictor effect. OBJECTIVES We investigated UII levels, hypertension, and other atherosclerotic risk factors in patients with SCF, a variety of coronary artery disease. METHODS We enrolled 14 patients with SCF and 29 subjects with normal coronary arteries without SCF. We compared the UII levels and the atherosclerotic risk factors between patients with SCF and control subjects with normal coronary flow. RESULTS UII concentrations were significantly higher in patients with SCF compared to controls (711.0 ± 19.4 vs. 701.5 ± 27.2 ng/mL, p = 0.006). We detected a positive correlation between SCF and age (r = 0.476, p = 0.001), BMI (r = 0.404, p = .002), UII concentrations (r = 0.422, p = 0.006), and hypertension (r = 0.594, p = 0.001). CONCLUSION We identified increased UII levels in patients with SCF. We think that UII concentrations may be informative on SCF pathogenesis due to relationship with inflammation, atherosclerosis, and vascular remodeling.
Collapse
Affiliation(s)
- Ömer Şatıroğlu
- Department of Cardiology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | - Mustafa Çetin
- Department of Cardiology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Yüksel Çiçek
- Department of Cardiology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Turan Erdoğan
- Department of Cardiology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Hakan Duman
- Department of Cardiology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| |
Collapse
|
8
|
Vanegas G, Leprince J, Lancien F, Mimassi N, Vaudry H, Le Mével JC. Divergent cardio-ventilatory and locomotor effects of centrally and peripherally administered urotensin II and urotensin II-related peptides in trout. Front Neurosci 2015; 9:142. [PMID: 25954149 PMCID: PMC4406059 DOI: 10.3389/fnins.2015.00142] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/06/2015] [Indexed: 12/13/2022] Open
Abstract
The urotensin II (UII) gene family consists of four paralogous genes called UII, UII-related peptide (URP), URP1 and URP2. UII and URP peptides exhibit the same cyclic hexapeptide core sequence (CFWKYC) while the N- and C-terminal regions are variable. UII, URP1, and URP2 mRNAs are differentially expressed within the central nervous system of teleost fishes, suggesting that they may exert distinct functions. Although the cardiovascular, ventilatory and locomotor effects of UII have been described in teleosts, much less is known regarding the physiological actions of URPs. The goal of the present study was to compare the central and peripheral actions of picomolar doses (5-500 pmol) of trout UII, URP1, and URP2 on cardio-ventilatory variables and locomotor activity in the unanesthetized trout. Compared to vehicle, intracerebroventricular injection of UII, URP1 and URP2 evoked a gradual increase in total ventilation (V TOT) reaching statistical significance for doses of 50 and 500 pmol of UII and URP1 but for only 500 pmol of URP2. In addition, UII, URP1 and URP2 provoked an elevation of dorsal aortic blood pressure (P DA) accompanied with tachycardia. All peptides caused an increase in locomotor activity (A CT), at a threshold dose of 5 pmol for UII and URP1, and 50 pmol for URP2. After intra-arterial (IA) injection, and in contrast to their central effects, only the highest dose of UII and URP1 significantly elevated V TOT and A CT. UII produced a dose-dependent hypertensive effect with concomitant bradycardia while URP1 increased P DA and heart rate after injection of only the highest dose of peptide. URP2 did not evoke any cardio-ventilatory or locomotor effect after IA injection. Collectively, these findings support the hypothesis that endogenous UII, URP1 and URP2 in the trout brain may act as neurotransmitters and/or neuromodulators acting synergistically or differentially to control the cardio-respiratory and locomotor systems. In the periphery, the only physiological actions of these peptides might be those related to the well-known cardiovascular regulatory actions of UII. It remains to determine whether the observed divergent physiological effects of UII and URPs are due to differential interaction with the UT receptor or binding to distinct UT subtypes.
Collapse
Affiliation(s)
- Gilmer Vanegas
- Institut National de la Santé et de la Recherche Médicale UMR1101, Laboratoire de Neurophysiologie, SFR ScInBioS, Université de Brest Brest, France
| | - Jérôme Leprince
- Institut National de la Santé et de la Recherche Médicale U982, UA Centre National de la Recherche Scientifique, Différenciation et Communication Neuronale et Neuroendocrine, Université de Rouen Mont-Saint-Aignan, France
| | - Frédéric Lancien
- Institut National de la Santé et de la Recherche Médicale UMR1101, Laboratoire de Neurophysiologie, SFR ScInBioS, Université de Brest Brest, France
| | - Nagi Mimassi
- Institut National de la Santé et de la Recherche Médicale UMR1101, Laboratoire de Neurophysiologie, SFR ScInBioS, Université de Brest Brest, France
| | - Hubert Vaudry
- Institut National de la Santé et de la Recherche Médicale U982, UA Centre National de la Recherche Scientifique, Différenciation et Communication Neuronale et Neuroendocrine, Université de Rouen Mont-Saint-Aignan, France
| | - Jean-Claude Le Mével
- Institut National de la Santé et de la Recherche Médicale UMR1101, Laboratoire de Neurophysiologie, SFR ScInBioS, Université de Brest Brest, France
| |
Collapse
|
9
|
Vaudry H, Leprince J, Chatenet D, Fournier A, Lambert DG, Le Mével JC, Ohlstein EH, Schwertani A, Tostivint H, Vaudry D. International Union of Basic and Clinical Pharmacology. XCII. Urotensin II, urotensin II-related peptide, and their receptor: from structure to function. Pharmacol Rev 2015; 67:214-58. [PMID: 25535277 DOI: 10.1124/pr.114.009480] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Urotensin II (UII) is a cyclic neuropeptide that was first isolated from the urophysis of teleost fish on the basis of its ability to contract the hindgut. Subsequently, UII was characterized in tetrapods including humans. Phylogenetic studies and synteny analysis indicate that UII and its paralogous peptide urotensin II-related peptide (URP) belong to the somatostatin/cortistatin superfamily. In mammals, the UII and URP genes are primarily expressed in cholinergic neurons of the brainstem and spinal cord. UII and URP mRNAs are also present in various organs notably in the cardiovascular, renal, and endocrine systems. UII and URP activate a common G protein-coupled receptor, called UT, that exhibits relatively high sequence identity with somatostatin, opioid, and galanin receptors. The UT gene is widely expressed in the central nervous system (CNS) and in peripheral tissues including the retina, heart, vascular bed, lung, kidney, adrenal medulla, and skeletal muscle. Structure-activity relationship studies and NMR conformational analysis have led to the rational design of a number of peptidic and nonpeptidic UT agonists and antagonists. Consistent with the wide distribution of UT, UII has now been shown to exert a large array of biologic activities, in particular in the CNS, the cardiovascular system, and the kidney. Here, we review the current knowledge concerning the pleiotropic actions of UII and discusses the possible use of antagonists for future therapeutic applications.
Collapse
Affiliation(s)
- Hubert Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jérôme Leprince
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Chatenet
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Alain Fournier
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David G Lambert
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jean-Claude Le Mével
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Eliot H Ohlstein
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Adel Schwertani
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Hervé Tostivint
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| |
Collapse
|
10
|
Brailoiu GC, Deliu E, Rabinowitz JE, Tilley DG, Koch WJ, Brailoiu E. Urotensin II promotes vagal-mediated bradycardia by activating cardiac-projecting parasympathetic neurons of nucleus ambiguus. J Neurochem 2014; 129:628-36. [PMID: 24521102 DOI: 10.1111/jnc.12679] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/31/2014] [Accepted: 02/05/2014] [Indexed: 12/31/2022]
Abstract
Urotensin II (U-II) is a cyclic undecapeptide that regulates cardiovascular function at central and peripheral sites. The functional role of U-II nucleus ambiguus, a key site controlling cardiac tone, has not been established, despite the identification of U-II and its receptor at this level. We report here that U-II produces an increase in cytosolic Ca(2+) concentration in retrogradely labeled cardiac vagal neurons of nucleus ambiguus via two pathways: (i) Ca(2+) release from the endoplasmic reticulum via inositol 1,4,5-trisphosphate receptor; and (ii) Ca(2+) influx through P/Q-type Ca(2+) channels. In addition, U-II depolarizes cultured cardiac parasympathetic neurons. Microinjection of increasing concentrations of U-II into nucleus ambiguus elicits dose-dependent bradycardia in conscious rats, indicating the in vivo activation of the cholinergic pathway controlling the heart rate. Both the in vitro and in vivo effects were abolished by the urotensin receptor antagonist, urantide. Our findings suggest that, in addition, to the previously reported increase in sympathetic outflow, U-II activates cardiac vagal neurons of nucleus ambiguus, which may contribute to cardioprotection.
Collapse
Affiliation(s)
- Gabriela Cristina Brailoiu
- Department of Pharmaceutical Sciences, Thomas Jefferson University, Jefferson School of Pharmacy, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
Renal expression of the peptide hormone urotensin II (UII) and its receptor (UTR) are dependent on kidney maturation and anatomical regions. However, renal regional hemodynamic effects of UII in neonates are unclear. Here, we investigated regional hemodynamic responses to acute intrarenal arterial administration of UII in newborn pigs. Western immunoblotting and immunofluorescence confirmed UTR expression and membrane localization in newborn pig renal afferent arterioles and afferent arteriolar smooth muscle cells respectively. Intrarenal arterial bolus injections of human UII (hUII; 1-100 ng/kg) resulted in a dose-dependent decrease in total renal blood flow (RBF) and an increase in mean arterial pressure (MAP) and renal vascular resistance (RVR) in newborn pigs. Moreover, hUII dose dependently reduced cortical blood flow (CBF) but increased medullary blood flow (MBF) in the piglets. hUII-induced MAP elevation and hemodynamic changes were inhibited by urantide, a UTR antagonist, but not losartan, a type 1 angiotensin II receptor antagonist. U-73122, a phospholipase C (PLC) inhibitor, and 2-aminoethoxydiphenyl borate, an inositol 1,4,5 trisphosphate (IP₃) receptor antagonist, attenuated hUII-induced MAP and RVR elevations, RBF and CBF reductions, but not MBF increase. These findings indicate that intrarenal arterial administration of hUII elevates blood pressure and induces region-selective renal hemodynamic changes in newborn pigs. Our data also suggest that the PLC/IP₃ signaling pathway contributes to hUII-induced alterations in MAP, RBF, RVR, and CBF but not MBF in newborn pigs.
Collapse
Affiliation(s)
- Hitesh Soni
- Department of Physiology, , University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | |
Collapse
|
12
|
Vaudry H, Do Rego JC, Le Mevel JC, Chatenet D, Tostivint H, Fournier A, Tonon MC, Pelletier G, Conlon JM, Leprince J. Urotensin II, from fish to human. Ann N Y Acad Sci 2010; 1200:53-66. [PMID: 20633133 DOI: 10.1111/j.1749-6632.2010.05514.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The cyclic peptide urotensin II (UII) was originally isolated from the urophysis of teleost fish on the basis of its ability to contract intestinal smooth muscle. The UII peptide has subsequently been isolated from frog brain and, later on, the pre-proUII cDNA has been characterized in mammals, including humans. A UII paralog called urotensin II-related peptide (URP) has been identified in the rat brain. The UII and URP genes originate from the same ancestral gene as the somatostatin and cortistatin genes. In the central nervous system (CNS) of tetrapods, UII is expressed primarily in motoneurons of the brainstem and spinal cord. The biological actions of UII and URP are mediated through a G protein-coupled receptor, termed UT, that exhibits high sequence similarity with the somatostatin receptors. The UT gene is widely expressed in the CNS and in peripheral organs. Consistent with the broad distribution of UT, UII and URP exert a large array of behavioral effects and regulate endocrine, cardiovascular, renal, and immune functions.
Collapse
Affiliation(s)
- Hubert Vaudry
- Laboratory of Cellular Neuroendocrinology, INSERM U413, European Institute for Peptide Research (IFRMP 23), University of Rouen, Mont-Saint-Aignan, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ross B, McKendy K, Giaid A. Role of urotensin II in health and disease. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1156-72. [DOI: 10.1152/ajpregu.00706.2009] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) is an 11 amino acid cyclic peptide originally isolated from the goby fish. The amino acid sequence of UII is exceptionally conserved across most vertebrate taxa, sharing structural similarity to somatostatin. UII binds to a class of G protein-coupled receptor known as GPR14 or the urotensin receptor (UT). UII and its receptor, UT, are widely expressed throughout the cardiovascular, pulmonary, central nervous, renal, and metabolic systems. UII is generally agreed to be the most potent endogenous vasoconstrictor discovered to date. Its physiological mechanisms are similar in some ways to other potent mediators, such as endothelin-1. For example, both compounds elicit a strong vascular smooth muscle-dependent vasoconstriction via Ca2+ release. UII also exerts a wide range of actions in other systems, such as proliferation of vascular smooth muscle cells, fibroblasts, and cancer cells. It also 1) enhances foam cell formation, chemotaxis of inflammatory cells, and inotropic and hypertrophic effects on heart muscle; 2) inhibits insulin release, modulates glomerular filtration, and release of catecholamines; and 3) may help regulate food intake and the sleep cycle. Elevated plasma levels of UII and increased levels of UII and UT expression have been demonstrated in numerous diseased conditions, including hypertension, atherosclerosis, heart failure, pulmonary hypertension, diabetes, renal failure, and the metabolic syndrome. Indeed, some of these reports suggest that UII is a marker of disease activity. As such, the UT receptor is emerging as a promising target for therapeutic intervention. Here, a concise review is given on the vast physiologic and pathologic roles of UII.
Collapse
Affiliation(s)
- Bryan Ross
- McGill University Health Center, Montreal, Quebec, Canada
| | | | - Adel Giaid
- McGill University Health Center, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Hunt BD, Ng LL, Lambert DG. A rat brain atlas of urotensin-II receptor expression and a review of central urotensin-II effects. Naunyn Schmiedebergs Arch Pharmacol 2010; 382:1-31. [PMID: 20422157 DOI: 10.1007/s00210-010-0503-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 02/22/2010] [Indexed: 02/07/2023]
Abstract
Urotensin-II (U-II) is an 11-amino acid cyclic peptide which exerts its actions through a G(q) protein-coupled receptor, UT. Much of the research focus of U-II is as a peptide of the periphery, particularly cardiovascular. Despite this, U-II was originally identified as a neuropeptide, and its expression is broad throughout the central nervous system. This brief review article catalogs the known sites of expression of UT within the CNS in the form of a diagrammatic rat brain atlas. Furthermore, the functional consequences of UT activation within specific brain regions are discussed along with the likely actions of synthetic UT ligands. Areas of high, medium, and low expression include the arcuate, paraventricular, and pedunculopontine tegmental nuclei, respectively. In the arcuate and paraventricular nuclei, where expression is high and moderate, U-II produces a pressor/tachycardic response in the former and a weaker response in the latter. Based on the known pharmacology of UT ligands (and assuming density is the primary determinant of efficacy in this case), we predict a weak response in the arcuate nucleus and possible antagonism of endogenous U-II response in the paraventricular nucleus by a low-efficacy partial agonist. These predicted responses lend themselves to relatively simple experimental verification.
Collapse
Affiliation(s)
- Benjamin D Hunt
- University Department of Cardiovascular Sciences (Pharmacology and Therapeutics Group), Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Leicester Royal Infirmary, Leicester LE1 5WW, UK
| | | | | |
Collapse
|
15
|
Sidharta PN, van Giersbergen PLM, Dingemanse J. Pharmacokinetics and pharmacodynamics of the urotensin-II receptor antagonist palosuran in healthy male subjects. J Clin Pharmacol 2009; 49:1168-75. [PMID: 19625629 DOI: 10.1177/0091270009341181] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Palosuran is a new potent and specific antagonist of the human urotensin II (U-II) receptor (UT receptor). This entry-into-humans study evaluated the tolerability and safety, pharmacokinetics, and pharmacodynamics of palosuran in a double-blind, placebo-controlled, single ascending-dose design. Oral doses of 5 to 2000 mg were given to 9 sequential groups of 8 healthy young men (6 on active drug, 2 on placebo) each. At regular intervals, tolerability and safety parameters and plasma levels of palosuran and U-II were determined. Urine was collected to determine excretion of sodium, potassium, creatinine, and palosuran. In this study, palosuran was well tolerated. No serious adverse events or dose-related adverse events were reported. No treatment-related pattern was detected for vital signs, clinical laboratory parameters, or electrocardiography parameters. After rapid absorption, palosuran displayed a plasma concentration-time profile characterized by 2 peaks at approximately 1 and 4 hours after drug administration. The apparent terminal elimination half-life was approximately 20 hours. AUC and C(max) values increased proportionally with doses up to 500 mg. Excretion of unchanged palosuran in urine was limited. No consistent effect was found on any of the pharmacodynamic variables measured. The results of this entry-into-humans study warrant further investigation of the therapeutic potential of palosuran.
Collapse
Affiliation(s)
- Patricia N Sidharta
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, CH-4123 Allschwil/Switzerland.
| | | | | |
Collapse
|
16
|
The Effects of Benzodiazepines on Urotensin II-Stimulated Norepinephrine Release from Rat Cerebrocortical Slices. Anesth Analg 2009; 108:1177-81. [DOI: 10.1213/ane.0b013e3181981faa] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Dubessy C, Cartier D, Lectez B, Bucharles C, Chartrel N, Montero-Hadjadje M, Bizet P, Chatenet D, Tostivint H, Scalbert E, Leprince J, Vaudry H, Jégou S, Lihrmann I. Characterization of urotensin II, distribution of urotensin II, urotensin II-related peptide and UT receptor mRNAs in mouse: evidence of urotensin II at the neuromuscular junction. J Neurochem 2008; 107:361-74. [PMID: 18710417 DOI: 10.1111/j.1471-4159.2008.05624.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Urotensin II (UII) and UII-related peptide (URP) are paralog neuropeptides whose existence and distribution in mouse have not yet been investigated. In this study, we showed by HPLC/RIA analysis that the UII-immunoreactive molecule in the mouse brain corresponds to a new UII(17) isoform. Moreover, calcium mobilization assays indicated that UII(17) and URP were equally potent in stimulating UII receptor (UT receptor). Quantitative RT-PCR and in situ hybridization analysis revealed that in the CNS UII and URP mRNAs were predominantly expressed in brainstem and spinal motoneurons. Besides, they were differentially expressed in the medial vestibular nucleus, locus coeruleus and the ventral medulla. In periphery, both mRNAs were expressed in skeletal muscle, testis, vagina, stomach, and gall bladder, whereas only URP mRNA could be detected in the seminal vesicle, heart, colon, and thymus. By contrast, the UT receptor mRNA was widely expressed, and notably, very high amounts of transcript occurred in skeletal muscle and prostate. In the biceps femoris muscle, UII-like immunoreactivity was shown to coexist with synaptophysin in muscle motor end plate regions. Altogether these results suggest that (i) UII and URP may have many redundant biological effects, especially at the neuromuscular junction; (ii) URP may more specifically participate to autonomic, cardiovascular and reproductive functions.
Collapse
Affiliation(s)
- Christophe Dubessy
- Neuronal and Neuroendocrine Communication and Differentiation, EA4310, INSERM U413, European Institute for Peptide Research (IFRMP 23), University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Urotensin II was first identified over 30 years ago as a potent vasoconstrictor, and the identification of its receptor in the heart, lungs, blood vessels, and brain have made it a potential target for human pharmacotherapy. Current research would suggest that urotensin II plays a major role in the pathophysiology of various cardiovascular disease entities. This article discusses the biologic effects of urotensin under normal and pathophysiologic conditions, and reviews the research experiences with synthetic urotensin blockers in the treatment of various cardiovascular illnesses.
Collapse
|
19
|
do Rego JC, Leprince J, Scalbert E, Vaudry H, Costentin J. Behavioral actions of urotensin-II. Peptides 2008; 29:838-44. [PMID: 18294732 DOI: 10.1016/j.peptides.2007.12.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2007] [Revised: 12/20/2007] [Accepted: 12/21/2007] [Indexed: 02/07/2023]
Abstract
Urotensin-II (U-II) and urotensin-II-related peptide (URP) have been identified as the endogenous ligands of the orphan G-protein-coupled receptor GPR14 now renamed UT. The occurrence of U-II and URP in the central nervous system, and the widespread distribution of UT in the brain suggest that U-II and URP may play various behavioral activities. Studies conducted in rodents have shown that central administration of U-II stimulates locomotion, provokes anxiety- and depressive-like states, enhances feeding activity and increases the duration of paradoxical sleep episodes. These observations indicate that, besides the endocrine/paracrine activities of U-II and URP on cardiovascular and kidney functions, these peptides may act as neurotransmitters and/or neuromodulators to regulate various neurobiological activities.
Collapse
Affiliation(s)
- Jean-Claude do Rego
- CNRS FRE 2735, Laboratoire de Neuropsychopharmacologie Expérimentale, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides 23, UFR de Médecine et Pharmacie, 22 Boulevard Gambetta, Rouen 76183, France.
| | | | | | | | | |
Collapse
|
20
|
Tölle M, van der Giet M. Cardiorenovascular effects of urotensin II and the relevance of the UT receptor. Peptides 2008; 29:743-63. [PMID: 17935830 DOI: 10.1016/j.peptides.2007.08.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Revised: 07/16/2007] [Accepted: 08/27/2007] [Indexed: 02/07/2023]
Abstract
Urotensin II (U-II) is a vasoactive peptide with many potent effects in the cardiorenovascular system. U-II activates a G-protein-coupled receptor termed UT. UT and U-II are highly expressed in the cardiovascular and renal system. Patients with various cardiovascular diseases show high U-II plasma levels. It was demonstrated that elevated U-II plasma levels and increased UT expression seem to play a role in heart failure, end-stage renal disease and atherosclerosis. U-II induces potent changes in vascular tone regulation. In addition, U-II stimulates vascular smooth muscle cell proliferation and cardiomyocyte hypertrophy. Currently several pharmaceutical companies are developing compounds to control the U-II/UT system. There are preclinical and some clinical studies showing potential benefits of inhibiting U-II function in renal disease, heart failure, and diabetes. This article will review both pre- and clinical data concerning cardiorenovascular effects of U-II.
Collapse
Affiliation(s)
- Markus Tölle
- Med. Klinik IV-Nephrology, Charite-Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany.
| | | |
Collapse
|
21
|
|
22
|
Watanabe T, Kanome T, Miyazaki A, Katagiri T. Human urotensin II as a link between hypertension and coronary artery disease. Hypertens Res 2006; 29:375-87. [PMID: 16940699 DOI: 10.1291/hypres.29.375] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hypertension is a well-known risk factor for atherosclerosis, but the molecular mechanisms that link elevated blood pressure to the progression of atherosclerosis remain unclear. Human urotensin II (U-II), the most potent endogenous vasoconstrictor peptide identified to date, and its receptor (UT receptor) are involved in the etiology of essential hypertension. In patients with essential hypertension, U-II infused into the forearm brachial artery has been shown to induce vasoconstriction. Recent studies have demonstrated elevated plasma U-II concentrations in patients with essential hypertension, diabetes mellitus, atherosclerosis, and coronary artery disease. U-II is expressed in endothelial cells, macrophages, macrophage-derived foam cells, and myointimal and medial vascular smooth muscle cells (VSMCs) of atherosclerotic human coronary arteries. UT receptors are present in VSMCs of human coronary arteries, the thoracic aorta and cardiac myocytes. Lymphocytes are the most active producers of U-II, whereas monocytes and macrophages are the major cell types expressing UT receptors, with relatively little receptor expression in foam cells, lymphocytes, and platelets. U-II accelerates foam cell formation by up-regulation of acyl-coenzyme A:cholesterol acyltransferase-1 in human monocyte-derived macrophages. In human endothelial cells, U-II promotes cell proliferation and up-regulates type 1 collagen expression. U-II also activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and plasminogen activator inhibitor-1 in human VSMCs, and stimulates VSMC proliferation with synergistic effects observed when combined with oxidized low-density lipoprotein, lysophosphatidylcholine, reactive oxygen species or serotonin. These findings suggest that U-II plays key roles in accelerating the development of atherosclerosis, thereby leading to coronary artery disease.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan.
| | | | | | | |
Collapse
|
23
|
Castel H, Diallo M, Chatenet D, Leprince J, Desrues L, Schouft MT, Fontaine M, Dubessy C, Lihrmann I, Scalbert E, Malagon M, Vaudry H, Tonon MC, Gandolfo P. Biochemical and functional characterization of high-affinity urotensin II receptors in rat cortical astrocytes. J Neurochem 2006; 99:582-95. [PMID: 16942596 DOI: 10.1111/j.1471-4159.2006.04130.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The urotensin II (UII) gene is primarily expressed in the central nervous system, but the functions of UII in the brain remain elusive. Here, we show that cultured rat astrocytes constitutively express the UII receptor (UT). Saturation and competition experiments performed with iodinated rat UII ([(125)I]rUII) revealed the presence of high- and low-affinity binding sites on astrocytes. Human UII (hUII) and the two highly active agonists hUII(4-11) and [3-iodo-Tyr9]hUII(4-11) were also very potent in displacing [(125)I]rUII from its binding sites, whereas the non-cyclic analogue [Ser5,10]hUII(4-11) and somatostatin-14 could only displace [(125)I]rUII binding at micromolar concentrations. Reciprocally, rUII failed to compete with [(125)I-Tyr0,D-Trp8]somatostatin-14 binding on astrocytes. Exposure of cultured astrocytes to rUII stimulated [(3)H]inositol incorporation and increased intracellular Ca(2+) concentration in a dose-dependent manner. The stimulatory effect of rUII on polyphosphoinositide turnover was abolished by the phospholipase C inhibitor U73122, but only reduced by 56% by pertussis toxin. The GTP analogue Gpp(NH)p caused its own biphasic displacement of [(125)I]rUII binding and provoked an affinity shift of the competition curve of rUII. Pertussis toxin shifted the competition curve towards a single lower affinity state. Taken together, these data demonstrate that rat astrocytes express high- and low-affinity UII binding sites coupled to G proteins, the high-affinity receptor exhibiting the same pharmacological and functional characteristics as UT.
Collapse
Affiliation(s)
- Hélène Castel
- INSERM, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Reyes BAS, Glaser JD, Magtoto R, Van Bockstaele EJ. Pro-opiomelanocortin colocalizes with corticotropin- releasing factor in axon terminals of the noradrenergic nucleus locus coeruleus. Eur J Neurosci 2006; 23:2067-77. [PMID: 16630054 DOI: 10.1111/j.1460-9568.2006.04744.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We previously demonstrated that the opioid peptide enkephalin and corticotropin-releasing factor (CRF) are occasionally colocalized in individual axon terminals but more frequently converge on common dendrites in the locus coeruleus (LC). To further examine potential opioid cotransmitters in CRF afferents we investigated the distribution of pro-opiomelanocortin (POMC), the precursor that yields the potent bioactive peptide beta-endorphin, with respect to CRF immunoreactivity using immunofluorescence and immunoelectron microscopic analyses of the LC. Coronal sections were collected through the dorsal pontine tegmentum of rat brain and processed for immunocytochemical detection of POMC and CRF or tyrosine hydroxylase (TH). POMC-immunoreactive processes exhibited a distinct distribution within the LC as compared to the enkephalin family of opioid peptides. Specifically, POMC fibers were enriched in the ventromedial aspect of the LC with fewer fibers present dorsolaterally. Immunofluorescence microscopy showed frequent coexistence of POMC and CRF in varicose processes that overlapped TH-containing somatodendritic processes in the LC. Ultrastructural analysis showed POMC immunoreactivity in unmyelinated axons and axon terminals. Axon terminals containing POMC were filled with numerous large dense-core vesicles. In sections processed for POMC and TH, approximately 29% of POMC-containing axon terminals (n = 405) targeted dendrites that exhibited immunogold-silver labeling for TH. In contrast, sections processed for POMC and CRF showed that 27% of POMC-labeled axon terminals (n = 657) also exhibited CRF immunoreactivity. Taken together, these data indicate that a subset of CRF afferents targeting the LC contain POMC and may be positioned to dually impact LC activity.
Collapse
Affiliation(s)
- Beverly A S Reyes
- Department of Neurosurgery, Further Institute for Neurosciences, Thomas Jefferson University, 900 Walnut Street, Suite 400, Philadelphia, PA 19107, USA.
| | | | | | | |
Collapse
|
25
|
Lacza Z, W Busija D. Urotensin-II is a nitric oxide-dependent vasodilator in the pial arteries of the newborn pig. Life Sci 2006; 78:2763-6. [PMID: 16337243 DOI: 10.1016/j.lfs.2005.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2005] [Accepted: 11/01/2005] [Indexed: 11/25/2022]
Abstract
Urotensin-II (UT-II) is a small circular peptide and is described as the most potent endogenous vasoconstrictor in various vascular beds. However, the in vivo effects of UT-II can be either vasoconstriction or vasodilation depending on the species and the tissue investigated. The present study sought to characterize the vasoactive effect of UT-II in the piglet cerebral circulation in vivo. Pial arteries of 99 +/- 6 microm were visualized with intravital microscopy through a closed cranial window in anesthetized newborn piglets. Topical application of UT-II elicited a weak dose-dependent vasodilation of the arteries (0.001 microM: 3 +/- 3 microm, 0.1 microM: 10 +/- 5 microm, 10 microM: 14 +/- 7 microm). Smaller arteries with an initial diameter below 100 microm showed minimal or no vasodilation, while larger arteries between 100 and 120 microm had a pronounced dose-dependent effect. Systemic application of 15 mg/kg Nomega-nitro-L-arginine-methyl ester (L-NAME) completely inhibited the vasodilation. We conclude that UT-II, in contrast to most other vascular beds, is a weak NO-dependent vasodilator in the piglet pial vasculature.
Collapse
Affiliation(s)
- Zsombor Lacza
- Department of Physiology/Pharmacology, Wake Forest University Health Sciences, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
26
|
Jégou S, Cartier D, Dubessy C, Gonzalez BJ, Chatenet D, Tostivint H, Scalbert E, LePrince J, Vaudry H, Lihrmann I. Localization of the urotensin II receptor in the rat central nervous system. J Comp Neurol 2006; 495:21-36. [PMID: 16432902 DOI: 10.1002/cne.20845] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The vasoactive peptide urotensin II (UII) is primarily expressed in motoneurons of the brainstem and spinal cord. Intracerebroventricular injection of UII provokes various behavioral, cardiovascular, motor, and endocrine responses in the rat, but the distribution of the UII receptor in the central nervous system (CNS) has not yet been determined. In the present study, we have investigated the localization of UII receptor (GPR14) mRNA and UII binding sites in the rat CNS. RT-PCR analysis revealed that the highest density of GPR14 mRNA occurred in the pontine nuclei. In situ hybridization histochemistry showed that the GPR14 gene is widely expressed in the brain and spinal cord. In particular, a strong hybridization signal was observed in the olfactory system, hippocampus, olfactory and medial amygdala, hypothalamus, epithalamus, several tegmental nuclei, locus coeruleus, pontine nuclei, motor nuclei, nucleus of the solitary tract, dorsal motor nucleus of the vagus, inferior olive, cerebellum, and spinal cord. Autoradiographic labeling of brain slices with radioiodinated UII showed the presence of UII-binding sites in the lateral septum, bed nucleus of the stria terminalis, medial amygdaloid nucleus, anteroventral thalamus, anterior pretectal nucleus, pedunculopontine tegmental nucleus, pontine nuclei, geniculate nuclei, parabigeminal nucleus, dorsal endopiriform nucleus, and cerebellar cortex. Intense expression of the GPR14 gene in some hypothalamic nuclei (supraoptic, paraventricular, ventromedian, and arcuate nuclei), in limbic structures (amygdala and hippocampus), in medullary nuclei (solitary tract, dorsal motor nucleus of the vagus), and in motor control regions (cerebral and cerebellar cortex, substantia nigra, pontine nuclei) provides the anatomical substrate for the central effects of UII on behavioral, cardiovascular, neuroendocrine, and motor functions. The occurrence of GPR14 mRNA in cranial and spinal motoneurons is consistent with the reported autocrine/paracrine action of UII on motoneurons.
Collapse
Affiliation(s)
- Sylvie Jégou
- Institut National de la Santé et de la Recherche Médicale U-413, Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research (IFRMP23), University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Huitron-Resendiz S, Kristensen MP, Sánchez-Alavez M, Clark SD, Grupke SL, Tyler C, Suzuki C, Nothacker HP, Civelli O, Criado JR, Henriksen SJ, Leonard CS, de Lecea L. Urotensin II modulates rapid eye movement sleep through activation of brainstem cholinergic neurons. J Neurosci 2006; 25:5465-74. [PMID: 15944374 PMCID: PMC6724976 DOI: 10.1523/jneurosci.4501-04.2005] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Urotensin II (UII) is a cyclic neuropeptide with strong vasoconstrictive activity in the peripheral vasculature. UII receptor mRNA is also expressed in the CNS, in particular in cholinergic neurons located in the mesopontine tegmental area, including the pedunculopontine tegmental (PPT) and lateral dorsal tegmental nuclei. This distribution suggests that the UII system is involved in functions regulated by acetylcholine, such as the sleep-wake cycle. Here, we tested the hypothesis that UII influences cholinergic PPT neuron activity and alters rapid eye movement (REM) sleep patterns in rats. Local administration of UII into the PPT nucleus increases REM sleep without inducing changes in the cortical blood flow. Intracerebroventricular injection of UII enhances both REM sleep and wakefulness and reduces slow-wave sleep 2. Intracerebroventricular, but not local, administration of UII increases cortical blood flow. Moreover, whole-cell recordings from rat-brain slices show that UII selectively excites cholinergic PPT neurons via an inward current and membrane depolarization that were accompanied by membrane conductance decreases. This effect does not depend on action potential generation or fast synaptic transmission because it persisted in the presence of TTX and antagonists of ionotropic glutamate, GABA, and glycine receptors. Collectively, these results suggest that UII plays a role in the regulation of REM sleep independently of its cerebrovascular actions by directly activating cholinergic brainstem neurons.
Collapse
|
28
|
Abstract
The discovery of novel biologically active peptides has led to an explosion in our understanding of the molecular mechanisms that underlie the regulation of sleep and wakefulness. Urotensin II (UII), a peptide originally isolated from fish and known for its strong cardiovascular effects in mammals, is another surprising candidate in the regulatory network of sleep. The UII receptor was found to be expressed by cholinergic neurons of laterodorsal and pedunculopontine tegmental nuclei, an area known to be of utmost importance for the on- and offset of rapid eye movement (REM) sleep. Recently, physiological data have provided further evidence that UII is indeed a modulator of REM sleep. The peptide directly excites cholinergic mesopontine neurons and increases the rate of REM sleep episodes. These new results and its emerging behavioral effects establish UII as a neurotransmitter/neuromodulator in mammals and should spark further interest into the neurobiological role of the peptide.
Collapse
Affiliation(s)
- Hans-Peter Nothacker
- Department of Pharmacology, University of California, Irvine, CA 92697-4625, USA.
| | | |
Collapse
|
29
|
Do-Rego JC, Chatenet D, Orta MH, Naudin B, Le Cudennec C, Leprince J, Scalbert E, Vaudry H, Costentin J. Behavioral effects of urotensin-II centrally administered in mice. Psychopharmacology (Berl) 2005; 183:103-17. [PMID: 16160878 DOI: 10.1007/s00213-005-0140-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Accepted: 07/21/2005] [Indexed: 11/28/2022]
Abstract
Urotensin-II (U-II) receptors are widely distributed in the central nervous system. Intracerebroventricular (i.c.v.) injection of U-II causes hypertension and bradycardia and stimulates prolactin and thyrotropin secretion. However, the behavioral effects of centrally administered U-II have received little attention. In the present study, we tested the effects of i.c.v. injections of U-II on behavioral, metabolic, and endocrine responses in mice. Administration of graded doses of U-II (1-10,000 ng/mouse) provoked: (1) a dose-dependent reduction in the number of head dips in the hole-board test; (2) a dose-dependent reduction in the number of entries in the white chamber in the black-and-white compartment test, and in the number of entries in the central platform and open arms in the plus-maze test; and (3) a dose-dependent increase in the duration of immobility in the forced-swimming test and tail suspension test. Intracerebroventricular injection of U-II also caused an increase in: food intake at doses of 100 and 1,000 ng/mouse, water intake at doses of 100-10,000 ng/mouse, and horizontal locomotion activity at a dose of 10,000 ng/mouse. Whatever was the dose, the central administration of U-II had no effect on body temperature, nociception, apomorphine-induced penile erection and climbing behavior, and stress-induced plasma corticosterone level. Taken together, the present study demonstrates that the central injection of U-II at doses of 1-10,000 ng/mouse induces anxiogenic- and depressant-like effects in mouse. These data suggest that U-II may be involved in some aspects of psychiatric disorders.
Collapse
Affiliation(s)
- Jean-Claude Do-Rego
- Institut Fédératif de Recherches Multidisciplinaires sur les Peptides (IFRMP 23), Laboratoire de Neuropsychopharmacologie Experimentale, CNRS FRE 2735, UFR de Medecine et Pharmacie, 76183 Rouen Cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Clark SD, Nothacker HP, Blaha CD, Tyler CJ, Duangdao DM, Grupke SL, Helton DR, Leonard CS, Civelli O. Urotensin II acts as a modulator of mesopontine cholinergic neurons. Brain Res 2005; 1059:139-48. [PMID: 16183039 DOI: 10.1016/j.brainres.2005.08.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2005] [Revised: 08/07/2005] [Accepted: 08/09/2005] [Indexed: 11/28/2022]
Abstract
Urotensin II (UII) is a vasomodulatory peptide that was not predicted to elicit CNS activity. However, because we have recently shown that the urotensin II receptor (UII-R) is selectively expressed in rat mesopontine cholinergic (MPCh) neurons, we hypothesize that UII may have a central function. The present study demonstrates that the UII system is able to modulate MPCh neuron activity. Brain slice experiments demonstrate that UII excites MPCh neurons of the mouse laterodorsal tegmentum (LDTg) by activating a slow inward current. Furthermore, microinfusion of UII into the ventral tegmental area produces a sustained increase in dopamine efflux in the nucleus accumbens, as measured by in vivo chronoamperometry. In agreement with UII activation of MPCh neurons, intracerebroventricular injections of UII significantly modulate ambulatory movements in both rats and mice but do not significantly affect startle habituation or prepulse inhibition. The present study establishes that UII is a neuromodulator that may be exploited to target disorders involving MPCh dysfunction.
Collapse
Affiliation(s)
- Stewart D Clark
- Department of Developmental and Cell Biology, University of California, Irvine, 92697-4625, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Urotensin II (U-II) is the most potent vasoconstrictor known, even more potent than endothelin-1. It was first isolated from the fish spinal cord and has been recognized as a hormone in the neurosecretory system of teleost fish for over 30 years. After the identification of U-II in humans and the orphan human G-protein-coupled receptor 14 as the urotensin II receptor, UT, many studies have shown that U-II may play an important role in cardiovascular regulation. Human urotensin II (hU-II) is an 11 amino acid cyclic peptide, generated by proteolytic cleavage from a precursor prohormone. It is expressed in the central nervous system as well as other tissues, such as kidney, spleen, small intestine, thymus, prostate, pituitary, and adrenal gland and circulates in human plasma. The plasma U-II level is elevated in renal failure, congestive heart failure, diabetes mellitus, systemic hypertension and portal hypertension caused by liver cirrhosis. The effect of U-II on the vascular system is variable, depending on species, vascular bed and calibre of the vessel. The net effect on vascular tone is a balance between endothelium-independent vasoconstriction and endothelium-dependent vasodilatation. U-II is also a neuropeptide and may play a role in tumour development. The development of UT receptor antagonists may provide a useful research tool as well as a novel treatment for cardiorenal diseases.
Collapse
Affiliation(s)
- Kwok Leung Ong
- Department of Medicine and the Research Centre of Heart, Brain, Hormone and Healthy Aging, University of Hong Kong, Hong Kong
| | | | | |
Collapse
|
32
|
Hood SG, Watson AMD, May CN. Cardiac actions of central but not peripheral urotensin II are prevented by beta-adrenoceptor blockade. Peptides 2005; 26:1248-56. [PMID: 15949643 DOI: 10.1016/j.peptides.2005.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 01/11/2005] [Accepted: 01/11/2005] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) is a highly conserved peptide that has potent cardiovascular actions following central and systemic administration. To determine whether the cardiovascular actions of UII are mediated via beta-adrenoceptors, we examined the effect of intravenous (IV) propranolol on the responses to intracerebroventricular (ICV) and IV administration of UII in conscious sheep. Sheep were surgically instrumented with ICV guide tubes and flow probes or cardiac sympathetic nerve recording electrodes. ICV UII (0.2 nmol/kg over 1 h) caused prolonged increases in heart rate (HR; 33 +/- 11 beats/min; P < 0.01), dF/dt (581 +/- 83 L/min/s; P < 0.001) and cardiac output (2.3 +/- 0.4 L/min; P < 0.001), accompanied by increases in coronary (19.8 +/- 5.4 mL/min; P < 0.01), mesenteric (211 +/- 50 mL/min; P < 0.05) and iliac (162 +/- 31 mL/min; P < 0.001) blood flows and plasma glucose (7.0 +/- 2.6 mmol/L; P < 0.05). Propranolol (30 mg bolus followed by 0.5 mg/kg/h IV) prevented the cardiac responses to ICV UII and inhibited the mesenteric vasodilatation. At 2 h after ICV UII, when HR and mean arterial pressure (MAP) were increased, cardiac sympathetic nerve activity (CSNA) was unchanged and the relation between CSNA and diastolic pressure was shifted to the right (P < 0.05). The hyperglycemia following ICV UII was abolished by ganglion blockade but not propranolol. IV UII (20 nmol/kg) caused a transient increase in HR and fall in stroke volume; these effects were not blocked by propranolol. These results demonstrate that the cardiac actions of central UII depend on beta-adrenoreceptor stimulation, secondary to increased CSNA and epinephrine release, whereas the cardiac actions of systemic UII are not mediated by beta-adrenoreceptors and probably depend on a direct action of UII on the heart.
Collapse
Affiliation(s)
- S G Hood
- Howard Florey Institute, University of Melbourne, Parkville, Vic. 3010, Australia
| | | | | |
Collapse
|
33
|
Doggrell SA. Urotensin-II and the cardiovascular system – the importance of developing modulators. Expert Opin Investig Drugs 2005; 13:479-87. [PMID: 15155123 DOI: 10.1517/13543784.13.5.479] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urotensin-II (U-II) potently contracts some large isolated blood vessels and cardiac tissue. However, the maximum effects on human blood vessels and heart are relatively small. U-II dilates human resistance arteries. It markedly decreased myocardial function and increased vascular resistance in cynomolgus monkeys, but the major effects of U-II have not been observed in healthy humans. A major role for U-II in human cardiovascular disease has not been clearly established despite studies in patients with coronary artery disease, heart failure, essential hypertension and diabetes. Peptide and non-peptide agonists and antagonists of the U-II receptor are being developed and will be useful in the characterisation of the effects of U-II, and may have some therapeutic potential.
Collapse
Affiliation(s)
- Sheila A Doggrell
- Doggrell Biomedical Communications, 47 Caronia Crescent, Lynfield, Auckland, New Zealand.
| |
Collapse
|
34
|
Horie S, Tsurumaki Y, Someya A, Hirabayashi T, Saito T, Okuma Y, Nomura Y, Murayama T. Involvement of cyclooxygenase-dependent pathway in contraction of isolated ileum by urotensin II. Peptides 2005; 26:323-9. [PMID: 15629545 DOI: 10.1016/j.peptides.2004.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2004] [Revised: 09/15/2004] [Accepted: 09/20/2004] [Indexed: 11/30/2022]
Abstract
We previously reported that urotensin II induced biphasic (brief- and long-lasting) contractions and the brief contraction was mediated by acetylcholine release from ganglionic cholinergic neurons in a segment of guinea-pig ileum. In the present work, we studied the mechanism contributing to long-lasting contractions induced by urotensin II. Treatment with 0.1 microM tetrodotoxin, 300 nM omega-conotoxin GVIA (an inhibitor of N-type Ca2+ channels) and 10 microM indomethacin (an inhibitor of cyclooxygenases) markedly inhibited 100 nM urotensin II-induced long-lasting contractions. The addition of 1 microM prostaglandin F2alpha (PGF2alpha) caused a limited brief contraction following long-lasting contraction, while 1 microM PGE2 induced marked biphasic contractions. Treatment with neurotoxins inhibited the long-lasting contractions induced by PGF2alpha and PGE2 without changing the PGE2-induced brief contractions. Treatment with 1 microM atropine markedly inhibited the urotensin II- and PGF2alpha-induced long-lasting contractions, but was less effective on the PGE2 responses. Treatment with a phospholipase A2 inhibitor decreased the urotensin II-induced contractions. These findings suggest that urotensin II induces, at least partially, long-lasting contractions via PG-sensitive cholinergic neurons and muscarinic acetylcholine receptors in the ileum.
Collapse
Affiliation(s)
- Syunji Horie
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Lin Y, Matsumura K, Tsuchihashi T, Fukuhara M, Fujii K, Iida M. Role of ERK and Rho kinase pathways in central pressor action of urotensin II. J Hypertens 2004; 22:983-8. [PMID: 15097239 DOI: 10.1097/00004872-200405000-00021] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND It has been shown that central urotensin II acts on the central nervous system to increase arterial pressure in conscious rats. OBJECTIVE To investigate the intracellular signal transduction mechanisms of the central cardiovascular action of urotensin II. METHODS The effects of intracerebroventricular (i.c.v.) administration of the extracellular signal-regulated protein kinase (ERK) inhibitor, PD 98059 (20 nmol), the phosphatidylinositol 3 (PI3) kinase inhibitor, wortmannin (20 nmol), or the Rho kinase inhibitor, Y-27632 (20 nmol), on cardiovascular responses to i.c.v. urotensin II (10 nmol) were determined in conscious rats. RESULTS I.c.v. injection of urotensin II increased both arterial pressure and heart rate (14.9 +/-1.5 mmHg and 94.6 +/-12.8 beats/min, respectively; P < 0.05 for each). Pretreatment with PD 98059 or Y-27632 significantly (P < 0.01 and P < 0.05, respectively) attenuated the pressor response induced by i.c.v. urotensin II (6.6 +/-1.4 and 6.9 +/-1.2 mmHg, respectively). Pretreatment with a mixed solution of PD 98059 and Y-27632 failed to cause further suppression of the urotensin II-induced pressor responses (4.5 +/-0.9 mmHg). In contrast, pretreatment with i.c.v. wortmannin failed to influence the pressor response induced by i.c.v. urotensin II (12.6 +/-1.3 mmHg). The tachycardiac response induced by i.c.v. urotensin II was not influenced by pretreatment with PD 98059, Y-27632 or wortmannin. CONCLUSIONS These findings suggest that the ERK and Rho kinase pathways, but not the PI3 pathway, may be involved in the central pressor action of urotensin II in conscious rats.
Collapse
Affiliation(s)
- Yingzi Lin
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Yu Y, Jawa A, Pan W, Kastin AJ. Effects of peptides, with emphasis on feeding, pain, and behavior A 5-year (1999-2003) review of publications in Peptides. Peptides 2004; 25:2257-89. [PMID: 15572212 DOI: 10.1016/j.peptides.2004.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Accepted: 09/21/2004] [Indexed: 11/28/2022]
Abstract
Novel effects of naturally occurring peptides are continuing to be discovered, and their mechanisms of actions as well as interactions with other substances, organs, and systems have been elucidated. Synthetic analogs may have actions similar or antagonistic to the endogenous peptides, and both the native peptides and analogs have potential as drugs or drug targets. The journal Peptides publishes many leading articles on the structure-activity relationship of peptides as well as outstanding reviews on some families of peptides. Complementary to the reviews, here we extract information from the original papers published during the past five years in Peptides (1999-2003) to summarize the effects of different classes of peptides, their modulation by other chemicals and various pathophysiological states, and the mechanisms by which the effects are exerted. Special attention is given to peptides related to feeding, pain, and other behaviors. By presenting in condensed form the effects of peptides which are essential for systems biology, we hope that this summary of existing knowledge will encourage additional novel research to be presented in Peptides.
Collapse
Affiliation(s)
- Yongmei Yu
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
37
|
Abstract
Urotensin II is a peptide present, together with its receptor, in the central nervous system and many peripheral tissues (including heart, blood vessels, kidneys and endocrine organs) of many species. The bioactive, mature form contains a cyclic heptapeptide perfectly preserved across species spanning 550 million years of evolution Its biological activity has been explored in cultured cells, in isolated vessels from several species, in the isolated perfused heart and in intact animals and man. Initial demonstration of potent vasoconstriction and cardiac depression by the human isoform in non-human primates has been followed by a series of reports indicating potent but highly variable and generally modest vascular responses dependent on species and vascular region. In man short term cardiovascular responses to administered urotensin II are small or absent. The place of urotensin II in the chronic trophic responses to cardiac and vascular injury and its possible roles as a neurotransmitter and/or regulator of renal and endocrine function remain largely unexplored.
Collapse
Affiliation(s)
- A Mark Richards
- Christchurch Cardioendocrine Research Group, Department of Medicine, Christchurch School of Medicine and Health Sciences, Riccarton Avenue, P.O. Box 4345, Christchurch, New Zealand.
| | | |
Collapse
|
38
|
Lancien F, Leprince J, Mimassi N, Mabin D, Vaudry H, Le Mével JC. Central effects of native urotensin II on motor activity, ventilatory movements, and heart rate in the trout Oncorhynchus mykiss. Brain Res 2004; 1023:167-74. [PMID: 15374742 DOI: 10.1016/j.brainres.2004.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2004] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) has been originally isolated from fish urophysis. However, in fish as in mammals, UII is also produced in brain neurons. Although UII binding sites are widely distributed in the fish central nervous system (CNS), little is known regarding its central activities. In the present study, we have investigated the effects of intracerebroventricular (ICV) administration of synthetic trout UII on the duration of motor activity (ACT; evidenced by bursts of activity on the trace of the ventilatory signal), ventilatory frequency (VF), ventilatory amplitude (VA), and heart rate (HR) in unanesthesized trout, Oncorhynchus mykiss. ICV injection of very low doses of UII (1 and 5 pmol) produced a dose-dependent increase of ACT without affecting VF, VA, or HR. At a higher dose (50 pmol), UII stimulated ACT as well as VF, VA, and HR. ICV injection of trout angiotensin II (5 pmol) did not affect ACT, VF, and VA, but provoked a robust increase in HR. These data provide the first evidence that central administration of UII stimulates motor activity in a nonmammalian vertebrate.
Collapse
Affiliation(s)
- Frédéric Lancien
- Laboratoire de Traitement de l'Information Médicale, INSERM U650, EA 2218, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest Cedex 3 29238, France
| | | | | | | | | | | |
Collapse
|
39
|
Watson AMD, May CN. Urotensin II, a novel peptide in central and peripheral cardiovascular control. Peptides 2004; 25:1759-66. [PMID: 15476943 DOI: 10.1016/j.peptides.2004.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Accepted: 04/15/2004] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) is a peptide that was originally isolated and characterized in fish. Interest in its effects in mammals increased with the identification of its receptor, G-protein coupled receptor 14, and its localization in humans. UII and its receptor have a wide distribution, including brain and spinal cord as well as heart, kidney and liver, implying that UII has important physiological actions. Recent studies suggest that UII may play an important role in the central nervous system. In conscious sheep, intracerebroventricular administration of UII induced large, prolonged increases in plasma epinephrine, adrenocorticotropic hormone, cardiac output and arterial pressure. Potent chronotropic and inotropic actions accompanied this, as well as peripheral vasodilatation. Administered intravenously, UII is an extremely potent vasoconstrictor in anesthetized monkeys, but reduces pressure in conscious and anesthetized rats, and causes a transient increase in conscious sheep, however vasomotor responses vary depending on species and vessel type. UII is elevated in conditions such as essential hypertension and heart failure suggesting a role in pathology. The results of studies with UII to date, together with its possible role in disease, emphasize the importance of examining the central and peripheral roles of UII in more detail.
Collapse
Affiliation(s)
- Anna M D Watson
- Howard Florey Institute, University of Melbourne, Parkville, Vic. 3010, Australia
| | | |
Collapse
|
40
|
Chatenet D, Dubessy C, Leprince J, Boularan C, Carlier L, Ségalas-Milazzo I, Guilhaudis L, Oulyadi H, Davoust D, Scalbert E, Pfeiffer B, Renard P, Tonon MC, Lihrmann I, Pacaud P, Vaudry H. Structure-activity relationships and structural conformation of a novel urotensin II-related peptide. Peptides 2004; 25:1819-30. [PMID: 15476952 DOI: 10.1016/j.peptides.2004.04.019] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2004] [Accepted: 04/23/2004] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) has been described as the most potent vasoconstrictor peptide and recognized as the endogenous ligand of the orphan G protein-coupled receptor GPR14. Recently, a UII-related peptide (URP) has been isolated from the rat brain and its sequence has been established as H-Ala-Cys-Phe-Trp-Lys-Tyr-Cys-Val-OH. In order to study the structure-function relationships of URP, we have synthesized a series of URP analogs and measured their binding affinity on hGPR14-transfected cells and their contractile activity in a rat aortic ring bioassay. Alanine substitution of each residue of URP significantly reduced the binding affinity and the contractile activity of the peptides, except for the Ala8-substituted analog that retained biological activity. Most importantly, D-scan of URP revealed that [D-Trp4]URP abrogated and [D-Tyr6]URP partially suppressed the UII-evoked contractile response. [Orn5]URP, which had very low agonistic efficacy, was the most potent antagonist in this series. The solution structure of URP has been determined by 1H NMR spectroscopy and molecular dynamics. URP exhibited a single conformation characterized by an inverse gamma-turn comprising residues Trp-Lys-Tyr which plays a crucial role in the biological activity of URP. These pharmacological and structural data should prove useful for the rational design of non-peptide ligands as potential GPR14 agonists and antagonists.
Collapse
Affiliation(s)
- David Chatenet
- Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research (IFRMP 23), INSERM U413, UA CNRS, University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Urotensin-II (UII) is a highly potent endogenous peptide within the cardiovascular system. Through stimulation of Galphaq-coupled UT receptors, UII mediates contraction of vascular smooth muscle and endothelial-dependent vasorelaxation, and positive inotropy in human right atrium and ventricle. A pathogenic role of the UT receptor system is emerging in cardiovascular disease states, with evidence for up-regulation of the UT receptor system in patients with congestive heart failure (CHF), pulmonary hypertension, cirrhosis and portal hypertension, and chronic renal failure. In vitro and in vivo studies show that under pathophysiological conditions, UII might contribute to cardiomyocyte hypertrophy, extracellular matrix production, enhanced vasoconstriction, vascular smooth muscle cell hyperplasia, and endothelial cell hyper-permeability. Single nucleotide polymorphisms of the UII gene may also impart a genetic predisposition of patients to diabetes. Therefore, the UT receptor system is a potential therapeutic target in the treatment of cardiac, pulmonary, and renal diseases. UT receptor antagonists are currently being developed to prevent and/or reverse the effects of over-activated UT receptors by the endogenous ligand. This review describes UII peptide and converting enzymes, and UT receptors in the cardiovascular system, focusing on pathophysiological roles of UII in the heart and blood vessels.
Collapse
Affiliation(s)
- Fraser D Russell
- Vascular Biology Laboratory, Department of Medicine, The University of Queensland, Brisbane, The Prince Charles Hospital, Pathology Building, Rode Road, Ground Floor, Room 3, Chermside 4032, Queensland, Australia.
| |
Collapse
|
42
|
Lin Y, Tsuchihashi T, Matsumura K, Fukuhara M, Ohya Y, Fujii K, Iida M. Central cardiovascular action of urotensin II in spontaneously hypertensive rats. Hypertens Res 2004; 26:839-45. [PMID: 14621188 DOI: 10.1291/hypres.26.839] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We have previously reported that urotensin II acts on the central nervous system to increase blood pressure in normotensive rats. In the present study, we have determined the central cardiovascular action of urotensin II in spontaneously hypertensive rats (SHR). Intracerebroventricular (ICV) injection of urotensin II elicited a dose-dependent increase in blood pressure in both SHR and normotensive Wistar-Kyoto rats (WKY). The changes in mean arterial pressure induced by ICV urotensin II at doses of 1 and 10 nmol in the WKY were 8 +/- 2 and 23 +/- 3 mmHg, respectively. ICV administration of urotensin II caused significantly greater increases in blood pressure in SHR (16 +/- 3 mmHg at 1 nmol and 35 +/- 3 mmHg at 10 nmol, respectively) compared with those in WKY. Urotensin II (10 nmol) elicited significant and comparable increases in heart rate in SHR (107 +/- 10 bpm) and WKY (101 +/- 21 bpm). Plasma epinephrine concentrations after ICV administration of 10 nmol urotensin II were 203 +/- 58 pmol/ml in SHR and 227 +/- 47 pmol/ml in WKY, which tended to be higher than those in artificial cerebrospinal fluid-injected rats (73+/- 7 and 87 +/- 28 pmol/ml, respectively, p < 0.1). The immunoreactivity of urotensin II receptor GPR 14 was expressed extensively in the glial cells within the brainstem, hypothalamus, and thalamus. These results suggest that central urotensin II may play a role in the pathogenesis of hypertension in SHR. Since GPR 14 was expressed in the glial cells of the brain, urotensin II may act as a neuromodulator to regulate blood pressure.
Collapse
Affiliation(s)
- Yingzi Lin
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Watson AMD, Lambert GW, Smith KJ, May CN. Urotensin II acts centrally to increase epinephrine and ACTH release and cause potent inotropic and chronotropic actions. Hypertension 2003; 42:373-9. [PMID: 12885791 DOI: 10.1161/01.hyp.0000084633.85427.e6] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urotensin II is a small peptide whose receptor was recently identified in mammals as the orphan G protein-coupled receptor-14. The reported cardiovascular responses to systemic urotensin II administration are variable, and there is little detailed information on its central cardiovascular actions. We examined the cardiovascular and humoral actions of intracerebroventricular urotensin II (0.02 and 0.2 nmol/kg and vehicle) and intravenous urotensin II (2, 20, and 40 nmol/kg and vehicle) in conscious ewes previously surgically implanted with flow probes and intracerebroventricular guide tubes. Two hours after intracerebroventricular infusion of urotensin II (0.2 nmol/kg over 1 hour; n=5), heart rate (+56+/-13 beats per minute [bpm]), dF/dt (an index of cardiac contractility; +533+/-128 L x min(-1) x s(-1)), and cardiac output (+3.4+/-0.4 L/min) increased significantly compared with vehicle, as did renal, mesenteric, and iliac blood flows and conductances. Plasma epinephrine, adrenocorticotropic hormone, and glucose levels also increased dramatically (+753+/-166 pg/mL, +14.3+/-3.5 pmol/L, and +7.0+/-1.4 mmol/L, respectively). All of these variables remained elevated for up to 4 hours after infusion. In contrast, 1 hour after intravenous urotensin II (40 nmol/kg bolus; n=6), a sustained tachycardia (+25+/-8 bpm) ensued, but cardiac output, cardiac contractility, total peripheral conductance, and plasma glucose levels did not change significantly. In summary, this is the first study to show that urotensin II acts centrally to stimulate sympathoadrenal and pituitary-adrenal pathways, resulting in increased adrenocorticotropic hormone and epinephrine release and potent chronotropic and inotropic actions. In contrast, tachycardia was the only major response to intravenous urotensin II. These findings suggest that urotensin II is a novel stimulator of central pathways that mediate responses to alerting stimuli or stress.
Collapse
Affiliation(s)
- Anna M D Watson
- Howard Florey Institute, University of Melbourne, Victoria 3010, Australia
| | | | | | | |
Collapse
|
44
|
Behm DJ, Harrison SM, Ao Z, Maniscalco K, Pickering SJ, Grau EV, Woods TN, Coatney RW, Doe CPA, Willette RN, Johns DG, Douglas SA. Deletion of the UT receptor gene results in the selective loss of urotensin-II contractile activity in aortae isolated from UT receptor knockout mice. Br J Pharmacol 2003; 139:464-72. [PMID: 12770952 PMCID: PMC1573852 DOI: 10.1038/sj.bjp.0705254] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
1 Urotensin-II (U-II) is among the most potent mammalian vasoconstrictors identified and may play a role in the aetiology of essential hypertension. Currently, only one mouse U-II receptor (UT) gene has been cloned. It is postulated that this protein is solely responsible for mediating U-II-induced vasoconstriction. 2 This hypothesis has been investigated in the present study, which assessed basal haemodynamics and vascular reactivity to hU-II in wild-type (UT((+/+))) and UT receptor knockout (UT((-/-))) mice. 3 Basal left ventricular end-diastolic and end-systolic volumes/pressures, stroke volumes, mean arterial blood pressures, heart rates, cardiac outputs and ejection fractions in UT((+/+)) mice and in UT((-/-)) mice were similar. 4 Relative to UT((+/+)) mouse isolated thoracic aorta, where hU-II was a potent spasmogen (pEC(50)=8.26+/-0.08) that evoked relatively little vasoconstriction (17+/-2% 60 mM KCl), vessels isolated from UT((-/-)) mice did not respond to hU-II. However, in contrast, the superior mesenteric artery isolated from both the genotypes did not contract in the presence of hU-II. Reactivity to unrelated vasoconstrictors (phenylephrine, endothelin-1, KCl) and endothelium-dependent/independent vasodilator agents (carbachol, sodium nitroprusside) was similar in the aorta and superior mesenteric arteries isolated from both the genotypes. 5 The present study is the first to directly link hU-II-induced vasoconstriction with the UT receptor. Deletion of the UT receptor gene results in loss of hU-II contractile action with no 'nonspecific' alterations in vascular reactivity. However, as might be predicted based on the limited contractile efficacy recorded in vitro, the contribution that hU-II and its receptor make to basal systemic haemodynamics appears to be negligible in this species.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/physiology
- Body Weight
- Gene Targeting
- Genotype
- Hemodynamics
- Humans
- In Vitro Techniques
- Male
- Mesenteric Artery, Superior/drug effects
- Mesenteric Artery, Superior/physiology
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Urotensins/metabolism
- Urotensins/pharmacology
- Urotensins/physiology
- Vasoconstriction/drug effects
- Vasoconstriction/physiology
- Vasoconstrictor Agents/pharmacology
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- David J Behm
- Department of Vascular Biology, Cardiovascular and Urogenital Diseases Center of Excellence for Drug Discovery, GlaxoSmithKline, King of Prussia, PA 19406-0939, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhu YC, Zhu YZ, Moore PK. Magnifying endoscopic observation of the gastric mucosa, particularly in patients with atrophic gastritis. Br J Pharmacol 1978; 148:884-901. [PMID: 16783414 PMCID: PMC1751922 DOI: 10.1038/sj.bjp.0706800] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The gastric mucosal surface was observed using the magnifying fibergastroscope (FGS-ML), and the fine gastric mucosal patterns, which were even smaller than one unit of gastric area, were examined at a magnification of about 30. For simplicification, we classified these patterns by magnifying endoscopy in the following ways; FP, FIP, FSP, SP and MP, modifying Yoshii's classification under the dissecting microscope. The FIP, which was found to have round and long elliptical gastric pits, is a new addition to our endoscopic classification. The relationship between the FIP and the intermediate zone was evaluated by superficial and histological studies of surgical and biopsy specimens. The width of the band of FIP seems to be related to the severity of atrophic gastritis. Also, the transformation of FP to FIP was assessed by comparing specimens taken from the resected and residual parts of the stomach, respectively. Moreover, it appears that severe gastritis occurs in the gastric mucosa which shows a FIP. Therefore, we consider that the FIP indicates the position of the atrophic border.
Collapse
Affiliation(s)
- Yi-Chun Zhu
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China.
| | | | | |
Collapse
|
46
|
Douglas SA, Dhanak D, Johns DG. Magnifying endoscopic observation of the gastric mucosa, particularly in patients with atrophic gastritis. Endoscopy 1978; 25:76-85. [PMID: 15102493 DOI: 10.1016/j.tips.2003.12.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gastric mucosal surface was observed using the magnifying fibergastroscope (FGS-ML), and the fine gastric mucosal patterns, which were even smaller than one unit of gastric area, were examined at a magnification of about 30. For simplicification, we classified these patterns by magnifying endoscopy in the following ways; FP, FIP, FSP, SP and MP, modifying Yoshii's classification under the dissecting microscope. The FIP, which was found to have round and long elliptical gastric pits, is a new addition to our endoscopic classification. The relationship between the FIP and the intermediate zone was evaluated by superficial and histological studies of surgical and biopsy specimens. The width of the band of FIP seems to be related to the severity of atrophic gastritis. Also, the transformation of FP to FIP was assessed by comparing specimens taken from the resected and residual parts of the stomach, respectively. Moreover, it appears that severe gastritis occurs in the gastric mucosa which shows a FIP. Therefore, we consider that the FIP indicates the position of the atrophic border.
Collapse
Affiliation(s)
- Stephen A Douglas
- Department of Vascular Biology, Cardiovascular and Urogenital Centre of Excellence for Drug Discovery, GlaxoSmithKline, PO Box 1539, 709 Swedeland Road, King of Prussia PA 19406-0939, USA.
| | | | | |
Collapse
|