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
|
Jopek K, Tyczewska M, Szyszka M, Blatkiewicz M, Jopek M, Malendowicz LK, Ruciński M. Impact of Classic Adrenal Secretagogues on mRNA Levels of Urotensin II and Its Receptor in Adrenal Gland of Rats. Int J Mol Sci 2023; 24:13412. [PMID: 37686217 PMCID: PMC10488159 DOI: 10.3390/ijms241713412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Urotensin 2 (Uts2) is a biologically active peptide involved in the regulation of a variety of physiological and pathophysiological processes. In both the human and rat adrenal gland, the expressions of the Uts2 gene and its receptor (Uts2r) have been described. This paper focuses on the description of the hormonal control of the mRNA levels of urotensin II and its receptor in the adrenal gland of the rat, both in vitro and in vivo. The initial in vitro experiments were carried out on freshly isolated rat adrenocortical cells and their primary culture. The obtained results indicated a stimulating PKA-independent effect of ACTH on the Uts2 mRNA level in the tested cells, with no changes in the Uts2r transcript. Subsequent in vivo experiments showed that ACTH-induced adrenal growth was accompanied by an elevated level of the Uts2 mRNA, with unchanged expression of Uts2r. In the other types of in vivo gland growth studied, enucleation-induced adrenal regeneration and compensatory growth of the gland, the mRNA levels of the studied genes showed no significant differences. The only exception was hemiadrenalectomy, which led to a significant increase in Uts2 mRNA expression level 24 h after surgery. In 12-week-old rats of both sexes, gonadectomy led to a significant increase in the level of Uts2 mRNA in the adrenal gland, an effect that was prevented by sex hormones' replacement. No changes in Uts2r transcript levels were observed under these conditions. Thus, this study suggests that the regulation of Uts2 and Uts2r mRNA levels differs significantly in the rat adrenal gland. While Uts2 transcript levels appear to be mainly dependent on ACTH action, Uts2r mRNA levels are not under the control of this hormone.
Collapse
|
3
|
Urotensin II Exerts Pressor Effects By Stimulating Renin And Aldosterone Synthase Gene Expression. Sci Rep 2017; 7:13876. [PMID: 29066763 PMCID: PMC5654760 DOI: 10.1038/s41598-017-12613-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/07/2017] [Indexed: 01/23/2023] Open
Abstract
We investigated the in vivo pressor effects of the potent vasoconstrictor Urotensin II (UII). We randomized normotensive Sprague-Dawley rats into 4 groups that received a 7-day UII infusion (cases) or vehicle (controls). Group 1 received normal sodium intake; Group 2 underwent unilateral nephrectomy and salt loading; Group 3 received spironolactone, besides unilateral nephrectomy and salt loading; Group 4 only received spironolactone. UII raised BP transiently after a lag phase of 12-36 hours in Group 1, and progressively over the week in Group 2. Spironolactone did not affect blood pressure, but abolished both pressor effects of UII in Group 3, and left blood pressure unaffected in Group 4. UII increased by 7-fold the renal expression of renin in Group 2, increased aldosterone synthase expression in the adrenocortical zona glomerulosa, and prevented the blunting of renin expression induced by high salt. UII raises BP transiently when sodium intake and renal function are normal, but progressively in salt-loaded uninephrectomized rats. Moreover, it increases aldosterone synthase and counteracts the suppression of renin induced by salt loading. This novel action of UII in the regulation of renin and aldosterone synthesis could play a role in several clinical conditions where UII levels are up-regulated.
Collapse
|
4
|
Matsumoto T, Watanabe S, Kobayashi S, Ando M, Taguchi K, Kobayashi T. Age-Related Reduction of Contractile Responses to Urotensin II Is Seen in Aortas from Wistar Rats but Not from Type 2 Diabetic Goto-Kakizaki Rats. Rejuvenation Res 2016; 20:134-145. [PMID: 27841739 DOI: 10.1089/rej.2016.1864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vascular dysfunction is a common finding in type 2 diabetes, although the response to urotensin II (UII), a potent vasoconstrictor peptide, remains unclear. We investigated whether a UII-induced contraction was increased in the aortas from type 2 diabetic Goto-Kakizaki (GK) rats at the chronic stage. At 36 or 37 weeks of age (older group), a UII-induced contraction was seen in GK rats and was reduced by a Rho kinase inhibitor or urotensin receptor (UT) antagonist, whereas UII failed to induce a contraction in aortas from age-matched Wistar rats. In UII-stimulated aortas, the expression of Rho kinases, Rho A, and phosphorylated myosin phosphatase target subunit 1 did not change between the two groups; however, phosphorylation of extracellular-regulated kinase 1/2 and p38 mitogen-activated protein kinase (MAPK) was greater in GK than in Wistar rats. Compared to intact aortas, UII-induced contractions were slightly, but not significantly, increased by endothelial denudation of the aortas of Wistar rats at 24 weeks of age. At 6 weeks of age (young group), the UII-induced contractions were seen in GK and Wistar groups. The total expression and the membrane-to-cytosol ratio of the UT protein slightly decreased in Wistar aortas with aging but not in GK aortas. These results demonstrate that the UII-induced contraction gradually decreased with aging in Wistar rats and was preserved in type 2 diabetes. Although alterations of UII-induced contractions during aging and type 2 diabetes may be associated with kinase activities (MAPKs or Rho kinase) or receptor profiles, further investigations are necessary to clarify the mechanisms.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Shun Watanabe
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Shota Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Makoto Ando
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University , Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
5
|
Albanese I, Daskalopoulou SS, Yu B, You Z, Genest J, Alsheikh-Ali A, Schwertani AG. The Urotensin II System and Carotid Atherosclerosis: A Role in Vascular Calcification. Front Pharmacol 2016; 7:149. [PMID: 27375483 PMCID: PMC4894881 DOI: 10.3389/fphar.2016.00149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND AIMS The aims of the present study were to determine the expression of urotensin II (UII), urotensin-II related peptide (URP), and their receptor (UT) in stable and unstable carotid atherosclerosis, and determine the effects of UII on human aortic smooth muscle cell (SMCs) calcification. METHODS AND RESULTS We examined UII, URP, and UT protein expression in 88 carotid endarterectomy specimens using immunohistochemistry. Expression of UII, URP, and UT was more evident in unstable compared to stable plaques (P < 0.05). Multivariate Spearman correlation analyses revealed significant positive correlations between UII, URP and UT overall staining and presence of calcification, severity of stenosis and inflammation (P < 0.05). Subjects undergoing carotid endarterectomy had significantly higher plasma UII levels, as assessed by ELISA, when compared with normolipidemic healthy control subjects (P < 0.05). Incubation of human aortic SMCs cultured in phosphate media with varying concentrations of UII resulted in a significant increase in calcium deposition and alkaline phosphatase activity. UII also significantly increased β-catenin translocation and expression of ALPL, BMP2, ON, and SOX9 (P < 0.05). Incubation of cells with phosphate medium alone increased the expression of the pre-UT and mature UT (P < 0.01), and addition of UII had a synergistic effect on pre-UT protein expression (P < 0.001) compared to phosphate medium alone. CONCLUSIONS Upregulation of UII, URP, and UT in unstable carotid endarterectomy plaques and plasma, and the stimulatory effect of UII on vascular smooth muscle cell calcification suggest that the UII system may play a role in the pathogenesis of vascular calcification and stability of atherosclerosis.
Collapse
Affiliation(s)
| | | | - Bin Yu
- Cardiology, McGill University Health Center Montreal, QC, Canada
| | - Zhipeng You
- Cardiology, McGill University Health Center Montreal, QC, Canada
| | - Jacques Genest
- Cardiology, McGill University Health Center Montreal, QC, Canada
| | - Alawi Alsheikh-Ali
- Cardiology, College of Medicine, Mohammed Bin Rashid University Dubai, UAE
| | | |
Collapse
|
6
|
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: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [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
|
7
|
Tostivint H, Ocampo Daza D, Bergqvist CA, Quan FB, Bougerol M, Lihrmann I, Larhammar D. Molecular evolution of GPCRs: Somatostatin/urotensin II receptors. J Mol Endocrinol 2014; 52:T61-86. [PMID: 24740737 DOI: 10.1530/jme-13-0274] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Somatostatin (SS) and urotensin II (UII) are members of two families of structurally related neuropeptides present in all vertebrates. They exert a large array of biological activities that are mediated by two families of G-protein-coupled receptors called SSTR and UTS2R respectively. It is proposed that the two families of peptides as well as those of their receptors probably derive from a single ancestral ligand-receptor pair. This pair had already been duplicated before the emergence of vertebrates to generate one SS peptide with two receptors and one UII peptide with one receptor. Thereafter, each family expanded in the three whole-genome duplications (1R, 2R, and 3R) that occurred during the evolution of vertebrates, whereupon some local duplications and gene losses occurred. Following the 2R event, the vertebrate ancestor is deduced to have possessed three SS (SS1, SS2, and SS5) and six SSTR (SSTR1-6) genes, on the one hand, and four UII (UII, URP, URP1, and URP2) and five UTS2R (UTS2R1-5) genes, on the other hand. In the teleost lineage, all these have been preserved with the exception of SSTR4. Moreover, several additional genes have been gained through the 3R event, such as SS4 and a second copy of the UII, SSTR2, SSTR3, and SSTR5 genes, and through local duplications, such as SS3. In mammals, all the genes of the SSTR family have been preserved, with the exception of SSTR6. In contrast, for the other families, extensive gene losses occurred, as only the SS1, SS2, UII, and URP genes and one UTS2R gene are still present.
Collapse
Affiliation(s)
- Hervé Tostivint
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Daniel Ocampo Daza
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Christina A Bergqvist
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Feng B Quan
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Marion Bougerol
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Isabelle Lihrmann
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| | - Dan Larhammar
- Evolution des Régulations EndocriniennesUMR 7221 CNRS and Muséum National d'Histoire Naturelle, Paris, FranceDepartment of NeuroscienceScience for Life Laboratory, Uppsala University, Uppsala, SwedenInserm U982Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Institute for Research and Innovation (IRIB), Rouen University, Mont-Saint-Aignan, France
| |
Collapse
|
8
|
Al Kindi H, Hafiane A, You Z, Albanese I, Pilote L, Genest J, Schwertani A. Circulating levels of the vasoactive peptide urotensin II in patients with acute coronary syndrome and stable coronary artery disease. Peptides 2014; 55:151-7. [PMID: 24642358 DOI: 10.1016/j.peptides.2014.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/26/2014] [Accepted: 03/09/2014] [Indexed: 12/26/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide with various roles in cardiovascular physiology and pathophysiology. There is an accumulating evidence implicating UII in atherosclerosis and coronary artery disease, making it an important target in acute coronary syndrome (ACS). In this study, we sought to determine the plasma levels of UII in ACS patients within 48 h of clinical presentation and after a 12-week recovery period. We compared them to patients with stable coronary artery disease (CAD) and a control group of normolipidemic subjects without known CAD. Using a highly sensitive ELISA technique, we measured plasma UII in 27 ACS patients, 26 stable CAD patients and 22 age-matched controls. ACS patients had significantly elevated plasma UII during the first 48 h of clinical presentation compared to stable CAD patients and controls. We also found significant positive correlations between UII and CRP and with triglycerides and a significant negative correlation between UII and EF. There was no correlation with LDL-C. In conclusion, plasma UII levels were elevated in patients with acute coronary syndrome, particularly immediately after clinical presentation. This suggests an upregulation of UII expression in ACS.
Collapse
Affiliation(s)
- Hamood Al Kindi
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Anouar Hafiane
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Zhipeng You
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Isabella Albanese
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Louise Pilote
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jacques Genest
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Adel Schwertani
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Brancaccio D, Limatola A, Campiglia P, Gomez-Monterrey I, Novellino E, Grieco P, Carotenuto A. Urantide Conformation and Interaction with the Urotensin-II Receptor. Arch Pharm (Weinheim) 2013; 347:185-92. [DOI: 10.1002/ardp.201300269] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/17/2013] [Accepted: 09/23/2013] [Indexed: 01/18/2023]
Affiliation(s)
- Diego Brancaccio
- Department of Pharmacy; University of Naples “Federico II”; Naples Italy
| | - Antonio Limatola
- Department of Pharmacy; University of Naples “Federico II”; Naples Italy
| | - Pietro Campiglia
- Department of Pharmacy; University of Salerno; Fisciano Salerno Italy
| | | | - Ettore Novellino
- Department of Pharmacy; University of Naples “Federico II”; Naples Italy
| | - Paolo Grieco
- Department of Pharmacy; University of Naples “Federico II”; Naples Italy
| | - Alfonso Carotenuto
- Department of Pharmacy; University of Naples “Federico II”; Naples Italy
| |
Collapse
|
10
|
New insight into the binding mode of peptides at urotensin-II receptor by Trp-constrained analogues of P5U and urantide. J Pept Sci 2013; 19:293-300. [DOI: 10.1002/psc.2498] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 01/25/2013] [Accepted: 01/27/2013] [Indexed: 11/07/2022]
|
11
|
Guo XH, Feng ZJ. Role of urotensin-Ⅱ in the pathogenesis of liver cirrhosis and portal hypertension and collateral circulation. Shijie Huaren Xiaohua Zazhi 2012; 20:3536-3541. [DOI: 10.11569/wcjd.v20.i35.3536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Urotensin-Ⅱ (U-Ⅱ) is a somatostatin-like cyclic peptide which has a potent vasoactive effect and can promote vascular reconstruction and hyperplasia. Research shows that UⅡ plays an important role in the development of liver cirrhosis and portal hypertension. UⅡ influences intrahepatic resistance and splanchnic hemodynamics through a variety of pathways, causing portal hypertension and participating in the formation of esophageal and gastric varices. UⅡ receptor antagonists can reduce portal pressure in cirrhotic rats, but this finding need to be confirmed clinically.
Collapse
|
12
|
Koulis C, de Haan JB, Allen TJ. Novel pathways and therapies in experimental diabetic atherosclerosis. Expert Rev Cardiovasc Ther 2012; 10:323-35. [PMID: 22390805 DOI: 10.1586/erc.12.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetic subjects are at a greater risk of developing major vascular complications due to abnormalities pertinent to the diabetic milieu. Current treatment options achieve significant improvements in glucose levels and blood pressure control, but do not necessarily prevent or retard diabetes-mediated macrovascular disease. In this review, we highlight several pathways that are increasingly being appreciated as playing a significant role in diabetic vascular injury. We focus particularly on the advanced glycation end product/receptor for advanced glycation end product (AGE/RAGE) axis and its interplay with the nuclear protein HMGB1. We discuss evidence implicating a significant role for the renin-angiotensin system, urotensin II and PPAR, as well as the importance of proinflammatory mediators and oxidative stress in cardiovascular complications. The specific targeting of these pathways may lead to novel therapies to reduce the burden of diabetic vascular complications.
Collapse
Affiliation(s)
- Christine Koulis
- Diabetic Complications Group, Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | |
Collapse
|
13
|
Nobata S, Donald JA, Balment RJ, Takei Y. Potent cardiovascular effects of homologous urotensin II (UII)-related peptide and UII in unanesthetized eels after peripheral and central injections. Am J Physiol Regul Integr Comp Physiol 2011; 300:R437-46. [DOI: 10.1152/ajpregu.00629.2010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We cloned cDNAs encoding urotensin II (UII)-related peptide (URP) and UII in Japanese eel, Anguilla japonica , the former being the first such cloning in teleost fishes. Unlike the exclusive expression of UII in the urophysis, the URP gene was expressed most abundantly in the brain (medulla oblongata) followed by the urophysis. Peripheral injections of URP into eels increased blood pressure by 16.1 ± 0.8 mmHg at 0.1 nmol/kg in ventral aortic blood pressure (PVA) and with similar potency and efficacy to that of UII (relative potency of URP to UII = 0.83). URP/UII and ANG II preferentially acted on the branchial and systemic circulations, respectively, and the duration of effect was distinct among the three peptides in the order of UII (60 min) >URP (30 min) >ANG II (14 min) in PVA. Urantide, a mammalian UII receptor antagonist, inhibited the URP effect (−63.6 ± 5.2%) to a greater extent than for UII (−39.9 ± 5.0%). URP and UII constricted isolated eel branchial and systemic arteries, showing their direct actions on the vascular smooth muscle. Central injection of URP increased blood pressure by 12.3 ± 0.8 mmHg at 50 pmol/eel in PVA and with similar efficacy but less potency (relative potency = 0.47) and shorter duration compared with UII. The central actions of URP/UII were more potent on the branchial circulation than on the systemic circulation, again opposite the effects of ANG II. The similar responses to peripheral and central injections suggest that peripheral hormones may act on the brain. Taken together, in eels, URP and UII are potent cardiovascular hormones like ANG II, acting directly on the peripheral vasculature, as well as a central vasomotor site, and their actions are mediated to different degrees by the UII receptor.
Collapse
Affiliation(s)
- Shigenori Nobata
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, University of Tokyo, Chiba, Japan
| | - John A. Donald
- School of Life and Environmental Sciences, Deakin University, Victoria, Australia; and
| | - Richard J. Balment
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Yoshio Takei
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, University of Tokyo, Chiba, Japan
| |
Collapse
|
14
|
Singh R, Rai U. Immunomodulatory role of urotensins in teleost Channa punctatus. Gen Comp Endocrinol 2011; 170:613-21. [PMID: 21130092 DOI: 10.1016/j.ygcen.2010.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 11/07/2010] [Accepted: 11/25/2010] [Indexed: 12/14/2022]
Abstract
The present study, for the first time in ectothermic vertebrates, reports the immunoregulatory role of urotensins I and II (UI and UII). Urotensins decreased the phagocytosis and nitrite production by splenic phagocytes. On superoxide production, UI had stimulatory while UII showed inhibitory effect. UI exerted its effect on phagocytes through corticotrophin-releasing factor (CRF) receptor as its non-specific antagonist astressin completely blocked the effect of UI on phagocytosis, nitrite release and superoxide production. Among the antagonists for specific CRF receptor 1 and 2, only CRF receptor 1 antagonist NBI 27914 abolished the effect of urotensin I. On the other hand, UII mediated its effect through urotensin receptor (UT receptor) since its antagonist urantide antagonized the effect of UII on phagocytosis, superoxide and nitrite release. These findings provide the direct evidence on physiological role of UI and UII through CRF receptor 1 and UT receptor, respectively in control of fish immune responses.
Collapse
Affiliation(s)
- Rajeev Singh
- Department of Zoology, University of Delhi, Delhi 110 007, India
| | | |
Collapse
|
15
|
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: 6.4] [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
|
16
|
Guidolin D, Albertin G, Ribatti D. Urotensin-II as an angiogenic factor. Peptides 2010; 31:1219-24. [PMID: 20346384 DOI: 10.1016/j.peptides.2010.03.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Revised: 03/17/2010] [Accepted: 03/17/2010] [Indexed: 02/07/2023]
Abstract
Angiogenesis, the process through which new blood vessels arise from pre-existing ones, is regulated by numerous "classic" factors and other "nonclassic" regulators of angiogenesis. Among these latter urotensin-II is a cyclic 11-amino acid (human) or 15-amino acid (rodent) peptide, originally isolated from the fish urophysis, which exerts a potent systemic vasoconstrictor and hypertensive effect. This review article summarizes the literature data concerning the involvement of urotensin-II in angiogenesis.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Human, Anatomy and Physiology (Section of Anatomy), University of Padova Medical School, Via Gabelli, 65, I-35121 Padova, Italy.
| | | | | |
Collapse
|
17
|
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: 7.6] [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
|
18
|
Grieco P, Carotenuto A, Campiglia P, Gomez-Monterrey I, Auriemma L, Sala M, Marcozzi C, d’Emmanuele di Villa Bianca R, Brancaccio D, Rovero P, Santicioli P, Meini S, Maggi CA, Novellino E. New Insight into the Binding Mode of Peptide Ligands at Urotensin-II Receptor: Structure−Activity Relationships Study on P5U and Urantide. J Med Chem 2009; 52:3927-40. [DOI: 10.1021/jm900148c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Paolo Grieco
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Alfonso Carotenuto
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Pietro Campiglia
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Isabel Gomez-Monterrey
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Luigia Auriemma
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Marina Sala
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Cristina Marcozzi
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Roberta d’Emmanuele di Villa Bianca
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Diego Brancaccio
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Paolo Rovero
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Paolo Santicioli
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Stefania Meini
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Carlo A. Maggi
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| | - Ettore Novellino
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Via D. Montesano, I-80131 Naples, Italy, Laboratorio Interdipartimentale di Chimica e Biologia dei Peptidi e Proteine, Department di Scienze Farmaceutiche, Università di Firenze, I-50019 Sesto Fiorentino, Florence, Italy, Department of Experimental Pharmacology, University of Naples “Federico II”, I-80131 Naples, Italy, Department of Pharmacology, Menarini Ricerche, Via Rismpondo 12/A, I-50131 Florence, Italy,
| |
Collapse
|
19
|
Zhang Y, Li Y, Wei R, Wang Z, Bu D, Zhao J, Pang Y, Tang C. Urotensin II is an autocrine/paracrine growth factor for aortic adventitia of rat. REGULATORY PEPTIDES 2008; 151:88-94. [PMID: 18955090 DOI: 10.1016/j.regpep.2008.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2008] [Revised: 09/07/2008] [Accepted: 10/01/2008] [Indexed: 02/05/2023]
Abstract
Urotensin II (UII) is a potent vasoconstrictive peptide; however, its significance in vascular adventitia has not been clearly elucidated. In this study, rat aortic adventitia showed mRNA expression and immunoreactivity of UII and its receptor (UT). Moreover, radioligand-binding assay showed that maximum binding capacity (Bmax) of [(125)I]-UII was higher in adventitia than in media (28.60+/-1.94 vs. 20.21+/-1.11 fmol/mg, P<0.01), with no difference in binding affinity (dissociation constant [Kd] 4.27+/-0.49 vs. 4.60+/-0.40 nM, P>0.05). Furthermore, in cultured adventitial fibroblasts, UII stimulated DNA synthesis, collagen synthesis and secretion in a concentration-dependent manner. These effects were inhibited by the UII receptor antagonist urantide (10(-6) mol/l), Ca(2+) channel blocker nicardipine (10(-5) mol/l), protein kinase C inhibitor H7 (10(-6) mol/l), and mitogen-activated protein kinase inhibitor PD98059 (10(-6) mol/l) but not the phosphatidyl inositol-3 kinase inhibitor wortmannin (10(-7) mol/l). UII may act as an autocrine/paracrine factor through its receptor and the Ca(2+) channel, protein kinase C, and mitogen-activated protein kinase signal transduction pathways, in the pathogenesis of vascular remodeling by activating vascular adventitia.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Cardiovascular Diseases, First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Pakala R. Role of urotensin II in atherosclerotic cardiovascular diseases. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2008; 9:166-78. [DOI: 10.1016/j.carrev.2008.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Revised: 01/24/2008] [Accepted: 02/05/2008] [Indexed: 02/07/2023]
|
21
|
Palosuran inhibits binding to primate UT receptors in cell membranes but demonstrates differential activity in intact cells and vascular tissues. Br J Pharmacol 2008; 155:374-86. [PMID: 18587423 DOI: 10.1038/bjp.2008.266] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The recent development of the UT ligand palosuran (1-[2-(4-benzyl-4-hydroxy-piperidin-1-yl)-ethyl]-3-(2-methyl-quinolin-4-yl)-urea sulphate salt) has led to the proposition that urotensin-II (U-II) plays a significant pathological role in acute and chronic renal injury in the rat. EXPERIMENTAL APPROACH In the present study, the pharmacological properties of palosuran were investigated further using a series of radioligand binding and functional bioassays. KEY RESULTS Palosuran functioned as a 'primate-selective' UT ligand in recombinant cell membranes (monkey and human UT K(i) values of 4 +/- 1 and 5 +/- 1 nM), lacking appreciable affinity at other mammalian UT isoforms (rodent and feline K(i) values >1 microM). Paradoxically, however, palosuran lost significant (10- to 54-fold) affinity for native and recombinant human UT when radioligand binding was performed in intact cells (K(i) values of 50 +/- 3 and 276 +/- 67 nM). In accordance, palosuran also exhibited diminished activity in hUT (human urotensin-II receptor)-CHO (Chinese hamster ovary) cells (IC50 323 +/- 67 nM) and isolated arteries (K(b)>10 microM in rat aorta; K(b)>8.5 microM in cat arteries; K(b)>1.6 microM in monkey arteries; K(b) 2.2 +/- 0.6 microM in hUT transgenic mouse aorta). Relative to recombinant binding K(i) values, palosuran was subjected to a 392- to 690-fold reduction in functional activity in monkey isolated arteries. Such phenomena were peculiar to palosuran and were not apparent with an alternative chemotype, SB-657510 (2-bromo-N-[4-chloro-3-((R)-1-methyl-pyrrolidin-3-yloxy)-phenyl]-4,5-dimethoxybenzenesulphonamide HCl). CONCLUSIONS AND IMPLICATIONS Collectively, such findings suggest that caution should be taken when interpreting data generated using palosuran. The loss of UT affinity/activity observed in intact cells and tissues cf. membranes offers a potential explanation for the disappointing clinical efficacy reported with palosuran in diabetic nephropathy patients. As such, the (patho)physiological significance of U-II in diabetic renal dysfunction remains uncertain.
Collapse
|
22
|
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
|
23
|
Lu W, Abdel-Razik AES, Ashton N, Balment RJ. Urotensin II: lessons from comparative studies for general endocrinology. Gen Comp Endocrinol 2008; 157:14-20. [PMID: 18440535 DOI: 10.1016/j.ygcen.2008.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 03/08/2008] [Accepted: 03/12/2008] [Indexed: 11/24/2022]
Abstract
The importance of combining studies across vertebrates to provide insights into the functionality of hormone systems is considered, using recent advances in Urotensin II (UII) biology to illustrate this. The impact of genome analyses on understanding ligand and UII receptor (UT) structures is reviewed, noting their high conservation from fish to mammals. The early linkage of UII with fish osmoregulatory physiology drove our investigation of possible renal actions of UII in mammals. The kidney is a potential major source of UII in mammals and endogenous peptide appears to have tonal influence over renal excretion of water and electrolytes. Blockade of UII actions by administration of UT receptor antagonist, urantide, in anaesthetised rats, indicates that endogenous UII lowers renal filtration rates and excretion of water and ions. These effects are considered in relation to apparent association of UII with a number of human cardiovascular and renal disorders. Following up the sequencing of UT in mammals here we contrast the first fish UT sequences with those in other species. It is now evident that UT expression in fish osmoregulatory tissues, such as the gill and kidney, exhibits considerable plasticity in response to physiological challenge, providing an important component of the adaptive organismal responses. A number of areas of UII research, which will continue to benefit from moving questions between appropriate vertebrate groups, have been highlighted. These comparative approaches will yield improved understanding and further novel actions of this intriguing endocrine and paracrine system, so highly conserved across the vertebrate series.
Collapse
Affiliation(s)
- W Lu
- Integrative Biology Division, Faculty of Life Sciences, University of Manchester, 3.614 Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | | | | | | |
Collapse
|
24
|
Vaudry H. Biologically active peptides urotensin II (UII) and urotensin II-related peptide (URP), and to their cognate receptor (UT). Editorial. Peptides 2008; 29:647-8. [PMID: 18314224 DOI: 10.1016/j.peptides.2008.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
25
|
Maguire JJ, Kuc RE, Kleinz MJ, Davenport AP. Immunocytochemical localization of the urotensin-II receptor, UT, to rat and human tissues: relevance to function. Peptides 2008; 29:735-42. [PMID: 17905478 DOI: 10.1016/j.peptides.2007.08.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 08/14/2007] [Accepted: 08/14/2007] [Indexed: 02/07/2023]
Abstract
We have examined whether differential expression of UT receptors in cardiovascular tissues from rats and humans may account for the diverse vascular actions reported for urotensin-II. We found UT immunoreactivity ubiquitously expressed in arterial and venous smooth muscle and cardiomyocytes in both species, however, compared to human, levels of UT immunoreactivity in rat vascular endothelial cells was below the level for detection. In rat skeletal muscle cells UT receptor localized to the sarcolemma, a pattern comparable to that for isoforms of nitric oxide synthase suggesting that urotensin-II mediated hindquarter vasodilatation may involve release of nitric oxide from skeletal muscle fibers.
Collapse
Affiliation(s)
- Janet J Maguire
- Clinical Pharmacology Unit, University of Cambridge, Level 6 Centre for Clinical Investigation, Box 110 Addenbrooke's Hospital, Cambridge CB2 2QQ, UK.
| | | | | | | |
Collapse
|
26
|
Le Mével JC, Lancien F, Mimassi N, Leprince J, Conlon JM, Vaudry H. Central and peripheral cardiovascular, ventilatory, and motor effects of trout urotensin-II in the trout. Peptides 2008; 29:830-7. [PMID: 17681644 DOI: 10.1016/j.peptides.2007.06.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 06/04/2007] [Accepted: 06/21/2007] [Indexed: 11/17/2022]
Abstract
Urotensin-II (U-II) was originally considered to be exclusively the product of the caudal neurosecretory system (CNSS) of teleost fish, but it has now been demonstrated that U-II is widely expressed in peripheral tissues and nervous structures of species from lampreys to mammals. However, very little is known regarding the physiological effects of this peptide in its species of origin. In the present review, we summarize the most significant results relating to the cardiovascular, ventilatory, and motor effects of centrally and peripherally administered synthetic trout U-II in our experimental animal model, the unanesthetized trout Oncorhynchus mykiss. In addition, we compare the actions of U-II with those of other neurohormonal peptides, particularly with the actions of urotensin-I, a 41-amino acid residue peptide paralogous to corticotropin-releasing hormone that is co-localized with U-II within neurons of the CNSS.
Collapse
Affiliation(s)
- Jean-Claude Le Mével
- INSERM U650, Laboratoire de Traitement de l'Information Médicale, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, 22 Avenue Camille Desmoulins, CS 93837, 29238 Brest Cedex 3, France.
| | | | | | | | | | | |
Collapse
|
27
|
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: 2.1] [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
|
28
|
Conlon JM. Liberation of urotensin II from the teleost urophysis: an historical overview. Peptides 2008; 29:651-7. [PMID: 17544546 DOI: 10.1016/j.peptides.2007.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 04/22/2007] [Accepted: 04/25/2007] [Indexed: 02/07/2023]
Abstract
During the past 20 years, urotensin II (UII) has progressed from being a peptide synthesized only in the urophysis of the caudal neurosecretory system of teleost fish to being considered an important physiological regulator in mammals with implications for the pathogenesis of a range of human cardiovascular and renal diseases. The "liberation" of UII from the urophysis was a gradual process and involved the sequential realization that (a) UII is present not only in the urophysis but also in the central nervous systems (CNS) of teleosts, (b) UII peptides, similar in structure to the urophysial peptides, are present in the diffuse caudal neurosecretory systems and/or CNS of species less evolutionarily advanced than teleosts, including Agnatha, thereby showing that UII is a phylogenetically ancient peptide, (c) UII is present in the brain and spinal cord of a tetrapod, the green frog Rana ridibunda, and (d) the UII gene and its specific receptor (GPR14/UT) are expressed in the CNS and certain peripheral tissues of mammals, including the human. The discovery that the genomes of mammals contain an additional gene encoding a UII-related peptide (URP) and the availability of highly effective peptide and non-peptide antagonists to investigate the role of UII in human physiology and pathophysiology ensure that the peptide will remain "center stage" for several years to come.
Collapse
Affiliation(s)
- J Michael Conlon
- Department of Biochemistry, Faculty of Medicine and Health Sciences, United Arab Emirates University, 17666 Al-Ain, United Arab Emirates.
| |
Collapse
|
29
|
|
30
|
McDonald J, Batuwangala M, Lambert DG. Role of urotensin II and its receptor in health and disease. J Anesth 2007; 21:378-89. [PMID: 17680191 DOI: 10.1007/s00540-007-0524-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Accepted: 03/15/2007] [Indexed: 02/07/2023]
Abstract
Urotensin II (U-II) is currently the most potent vasoconstrictor identified. This action is brought about via activation of a G(q/11)-protein coupled receptor (UT receptor). U-II activation of the UT receptor increases inositol phosphate turnover and intracellular Ca(2+). In addition to producing vasoconstriction, dilation and ionotropic effects have also been described. There is considerable variation in the responsiveness of particular vascular beds from the same and different species, including humans. Receptors for U-II are located peripherally on vascular smooth muscle (contractile responses) and endothelial cells (dilatory responses via nitric oxide). In humans, plasma U-II is elevated in heart failure, renal failure, liver disease, and diabetes. Iontophoresis of U-II in healthy volunteers produces vasodilation (of the forearm) while in patients with heart failure or hypertension a constriction is observed. To date there is only one clinical study using a UT receptor antagonist (palosuran) in diabetic patients with macroalbuminuria. This antagonist reduced albumin excretion, probably by increasing renal blood flow. Studies in other disease conditions are eagerly awaited. In summary, the U-II / UT receptor system has clinical potential, and for the anesthesiologist, this novel peptide-receptor system may be of use in the intensive care unit.
Collapse
Affiliation(s)
- John McDonald
- Department of Cardiovascular Sciences, Pharmacology and Therapeutics Group, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, LRI, Leicester, LE1 5WW, UK
| | | | | |
Collapse
|
31
|
Clark SD, Alderson HL, Winn P, Latimer MP, Nothacker HP, Civelli O. Fusion of diphtheria toxin and urotensin II produces a neurotoxin selective for cholinergic neurons in the rat mesopontine tegmentum. J Neurochem 2007; 102:112-20. [PMID: 17419804 DOI: 10.1111/j.1471-4159.2007.04529.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Urotensin II is a neuropeptide first isolated from fish and later found in mammals: where it has potent cardiovascular, endocrine and behavioral effects. In rat brain the urotensin II receptor (UII-R) is predominately expressed in the cholinergic neurons of the pedunculopontine (PPTg) and laterodorsal tegmental nuclei. Typically, the function of the PPTg has been examined using excitotoxins, destroying both cholinergic and non-cholinergic neurons, which confounds interpretation. We took advantage of UII-R's unique expression profile, by combining UII with diphtheria toxin, to engineer a toxin specific for cholinergic neurons of the PPTg. In vitro, two different toxin constructs were shown to selectively activate UII-R (average EC50 approximately 30 nmol/L; calcium mobility assay) and to be 10,000-fold more toxic to UII-R expressing CHO cells, than wildtype cells (average LD50 approximately 2 nmol/L; cell viability). In vivo, pressure injection into the PPTg of rats, resulted in specific loss of choline transporter and NADPH diaphorase positive neurons known to express the UII-R. The lesions developed over time, resulting in the loss of over 80% of cholinergic neurons at 21 days, with little damage to surrounding neurons. This is the first highly selective molecular tool for the depletion of mesopontine cholinergic neurons. The toxin will help to functionally dissect the pedunculopontine and laterodorsal tegmental nuclei, and advance the understanding of the functions of these structures.
Collapse
Affiliation(s)
- S D Clark
- Laboratory of Molecular Neurobiology, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
32
|
Chatenet D, Dubessy C, Boularan C, Scalbert E, Pfeiffer B, Renard P, Lihrmann I, Pacaud P, Tonon MC, Vaudry H, Leprince J. Structure-activity relationships of a novel series of urotensin II analogues: identification of a urotensin II antagonist. J Med Chem 2007; 49:7234-8. [PMID: 17125276 DOI: 10.1021/jm0602110] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urotensin II (U-II) is a potent vasoconstrictor peptide which has been identified as the endogenous ligand for the orphan G protein-coupled receptor GPR14 now renamed UT receptor. As the C-terminal cyclic hexapeptide of U-II (U-II(4-11), H-Asp-Cys-Phe-Trp-Lys-Tyr-Cys-Val-OH) possesses full biological activity, we have synthesized a series of U-II(4-11) analogues and measured their binding affinity on hGPR14-transfected CHO cells and their contractile activity on de-endothelialized rat aortic rings. The data indicate that a free amino group and a functionalized side-chain at the N-terminal extremity of the peptide are not required for biological activity. In addition, the minimal chemical requirement at position 9 of U-II(4-11) is the presence of an aromatic moiety. Most importantly, replacement of the Phe6 residue by cyclohexyl-Ala (Cha) led to an analogue, [Cha6]U-II(4-11), that was devoid of agonistic activity but was able to dose-dependently suppress the vasoconstrictor effect of U-II on rat aortic rings. These new pharmacological data, by providing further information regarding the structure-activity relationships of U-II analogues, should prove useful for the rational design of potent and nonpeptidic UT receptor agonists and antagonists.
Collapse
Affiliation(s)
- David Chatenet
- INSERM U413, Laboratory of Cellular & Molecular Neuroendocrinology, European Institute for Peptide Research (IFRMP 23), University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Behm DJ, Stankus G, Doe CPA, Willette RN, Sarau HM, Foley JJ, Schmidt DB, Nuthulaganti P, Fornwald JA, Ames RS, Lambert DG, Calo' G, Camarda V, Aiyar NV, Douglas SA. The peptidic urotensin-II receptor ligand GSK248451 possesses less intrinsic activity than the low-efficacy partial agonists SB-710411 and urantide in native mammalian tissues and recombinant cell systems. Br J Pharmacol 2006; 148:173-90. [PMID: 16547525 PMCID: PMC1617064 DOI: 10.1038/sj.bjp.0706716] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Several peptidic urotensin-II (UT) receptor antagonists exert 'paradoxical' agonist activity in recombinant cell- and tissue-based bioassay systems, likely the result of differential urotensin-II receptor (UT receptor) signal transduction/coupling efficiency between assays. The present study has examined this phenomenon in mammalian arteries and recombinant UT-HEK (human embryonic kidney) cells.BacMam-mediated recombinant UT receptor upregulation in HEK cells augmented agonist activity for all four peptidic UT ligands studied. The nominal rank order of relative intrinsic efficacy was U-II>urantide ([Pen(5)-DTrp(7)-Orn(8)]hU-II(4-11))>SB-710411 (Cpa-c[DCys-Pal-DTrp-Lys-Val-Cys]-Cpa-amide)>>GSK248451 (Cin-c[DCys-Pal-DTrp-Orn-Val-Cys]-His-amide) (the relative coupling efficiency of recombinant HEK cells was cat>human>>rat UT receptor). The present study further demonstrated that the use of high signal transduction/coupling efficiency isolated blood vessel assays (primate>cat arteries) is required in order to characterize UT receptor antagonism thoroughly. This cannot be attained simply by using the rat isolated aorta, an artery with low signal transduction/coupling efficiency in which low-efficacy agonists appear to function as antagonists. In contrast to the 'low-efficacy agonists' urantide and SB-710411, GSK248451 functioned as a potent UT receptor antagonist in all native isolated tissues studied (UT receptor selectivity was confirmed in the rat aorta). Further, GSK248451 exhibited an extremely low level of relative intrinsic activity in recombinant HEK cells (4-5-fold less than seen with urantide). Since GSK248451 (1 mg kg(-1), i.v.) blocked the systemic pressor actions of exogenous U-II in the anaesthetized cat, it represents a suitable peptidic tool antagonist for delineating the role of U-II in the aetiology of mammalian cardiometabolic diseases.
Collapse
Affiliation(s)
- David J Behm
- Department of Vascular Biology and Thrombosis, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Disa J, Floyd LE, Edwards RM, Douglas SA, Aiyar NV. Identification and characterization of binding sites for human urotensin-II in Sprague-Dawley rat renal medulla using quantitative receptor autoradiography. Peptides 2006; 27:1532-7. [PMID: 16290260 DOI: 10.1016/j.peptides.2005.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Revised: 10/05/2005] [Accepted: 10/05/2005] [Indexed: 02/07/2023]
Abstract
Urotensin-II (U-II), a ligand for the G-protein-coupled receptor UT, has been characterized as the most potent mammalian vasoconstrictor identified to date. Although circulating levels of U-II are altered in lower species (e.g., fish) upon exposure to hypo-osmotic stress, little is known about the actions of this cyclic undecapeptide within the kidney, an organ that plays a pivotal role in the control of cardiovascular homeostasis, influencing both cardiac preload (plasma volume) and after load (peripheral resistance). The present study reports the identification of specific, high affinity [125I]hU-II binding sites in Sprague-Dawley rat kidney outer medulla by autoradiography and also through membrane radioligand binding (Kd 1.9 +/- 0.9 nM and Bmax 408 +/- 47 amol mm(-2) and Kd 1.4 +/- 0.3 nM and Bmax 51.3 +/- 7.8 fmol mg(-1) protein, respectively). Differences were observed in the binding characteristics within rat strains. Compared to the Sprague-Dawley, Wistar Kyoto (WKY) and spontaneously hypertensive (SHR) rat kidney outer medulla displayed low density < 20 fmol mg(-1) protein and low affinity (> 1 microM) [125I]hU-II binding sites. Thus, the relative contribution of specific U-II binding sites to the physiological actions of U-II in the control of cardiorenal homeostasis is worthy of further investigation.
Collapse
Affiliation(s)
- Jyoti Disa
- Department of Vascular Biology and Thrombosis, Cardiovascular and Urogenital Center of Excellence for Drug Discovery, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | | | | | | | | |
Collapse
|
35
|
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
|
36
|
Song W, McDonald J, Camarda V, Calo G, Guerrini R, Marzola E, Thompson JP, Rowbotham DJ, Lambert DG. Cell and tissue responses of a range of Urotensin II analogs at cloned and native urotensin II receptors. Evidence for coupling promiscuity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2006; 373:148-57. [PMID: 16596397 DOI: 10.1007/s00210-006-0057-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Accepted: 03/01/2006] [Indexed: 10/24/2022]
Abstract
Urotensin II (U-II) is the peptide ligand for the G-protein-coupled U-II receptor (UT). U-II has been dubbed "the most potent vasoconstrictor identified to date". However, in vivo studies with this system are hampered by the paucity of available ligands. Here, we characterise Chinese hamster ovary (CHO) cells expressing the human UT receptor in the following assays; (1) [(125)I]U-II binding, (2) GTPgamma[(35)S] binding, (3) cAMP formation, and (4) intracellular Ca(2+). We assess activity of 9 U-II analogues using these paradigms and examine their ability to contract isolated rat aorta. CHO(hUT) cells bound [(125)I]U-II with a B (max) and K (d) of 1,110+/-70 fmol/mg protein and 742 pM, respectively. hU-II stimulated GTPgamma[(35)S] binding (pEC(50) 8.38), optimal at low (0.1 muM) GDP concentrations. The hU-II GTPgamma[(35)S] response was partially PTx sensitive and there was a potent (pEC(50) 9.23) low efficacy ( approximately 20% inhibition) coupling to adenylyl cyclase. In CHO(hUT) cells hU-II stimulates calcium release from intracellular stores (pEC(50) 8.80) and calcium influx in a PTx-insensitive manner. In our structure-activity relationship study most ligands acted as full agonists. However, urantide behaved as a partial agonist (pEC(50) 7.67/pK(B) 7.55) in GTPgamma[(35)S] binding, a full agonist (pEC(50) 8.11) for increases in intracellular Ca(2+) and a competitive antagonist in the rat aorta bioassay (pK(B) 8.59). Collectively, these data show promiscuity at high expression and indicate the need for careful multi-assay evaluation of novel U-II analogues. Further modification of urantide, in order to eliminate residual agonist activity and to identify novel ligands for in vivo cardiovascular studies are clearly warranted.
Collapse
Affiliation(s)
- Wei Song
- University Department of Cardiovascular Sciences (Pharmacology and Therapeutics Group), Division of Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, LE1 5WW, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wang ZJ, Shi LB, Xiong ZW, Zhang LF, Meng L, Bu DF, Tang CS, Ding WH. Alteration of vascular urotensin II receptor in mice with apolipoprotein E gene knockout. Peptides 2006; 27:858-63. [PMID: 16325305 DOI: 10.1016/j.peptides.2005.08.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Revised: 08/31/2005] [Accepted: 08/31/2005] [Indexed: 10/25/2022]
Abstract
Urotensin II (U-II), originally identified as a fish neuropeptide, exerts a broad spectrum of biological functions and is considered to be the most potent vasoconstrictor in mammals. Recently the intense staining of U-II-like immunoreactivity within coronary atheroma and the effect of mitogenic in vascular smooth muscle cells (VSMCs) suggest that U-II is possibly a proatherogenic factor in the pathogenesis of cardiovascular diseases (CVD). In the present study, we analyzed the gene expression of U-II and GPR14, a high-affinity receptor for U-II, in aorta of apolipoprotein E (apoE) -/- mutant mice by semi-quantitative reverse transcription polymerase chain reaction (RT-PCR). Compared with wild-type mice at the same age group, GPR14 mRNA level is significant increase in aorta of apoE -/- mice at age of 18, 28 and 38 weeks, respectively. The increased GPR14 mRNA level was 54.2, 50.0 and 97.0% in the three age groups, respectively. We did not detect U-II mRNA expression in aorta either in apoE -/- mice or in wild-type mice. In 28-week mice, ligand-binding assay showed that maximum binding capacity (Bmax) of 125I-U-II aorta from apoE -/- mice was increased by 64%, while the affinity of the binding did not change compared with that of wild-type mice. These results indicate that the up-regulation of vascular U-II receptor (UT), GPR14 might be involved in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Zhi Jian Wang
- Division of Cardiology, Department of Internal Medicine, Peking University First Hospital, Xishikudajie #8, West District, Beijing 10034, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Balment RJ, Song W, Ashton N. Urotensin II: Ancient Hormone with New Functions in Vertebrate Body Fluid Regulation. Ann N Y Acad Sci 2006; 1040:66-73. [PMID: 15891007 DOI: 10.1196/annals.1327.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Urotensin II (UII), described in many fish species, is secreted by the caudal neurosecretory system, a unique fish neuroendocrine structure. We have examined UII secretion and its control in euryhaline fish, supporting a proposed role in osmoregulation. However, it is now apparent that UII is present in other vertebrates, including mammals. The 12-amino-acid peptide has been highly conserved and the key cyclic region is common from fish to humans. Our UII radioimmunoassay for flounder, directed to this cyclic region, has shown circulating UII levels in humans and rats comparable with those in fish. In mammals, UII cardiovascular effects vary between species, with vasoconstriction only evident in specific vascular beds. The kidney expresses UII receptors and responds to UII administration by a reduction in glomerular filtration rate, urine flow, and excretion of the major ions. Interestingly, plasma levels of UII are chronically elevated in rat models of hypertension. These observations imply an unforeseen role for this ancient fish hormone in the physiological and perhaps pathophysiological regulation of body fluids in higher vertebrates, including humans.
Collapse
Affiliation(s)
- R J Balment
- Faculty of Life Sciences, University of Manchester, UK.
| | | | | |
Collapse
|
39
|
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
|
40
|
Gendron G, Gobeil F, Bélanger S, Gagnon S, Regoli D, D'Orléans-Juste P. Urotensin II-induced hypotensive responses in Wistar-Kyoto (Wky) and spontaneously hypertensive (Shr) rats. Peptides 2005; 26:1468-74. [PMID: 16042987 DOI: 10.1016/j.peptides.2005.03.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human urotensin II (hU-II) is a potent vasoactive peptide which modulates some of the functions of the cardiovascular and other systems. The in vivo mechanism of action by which hU-II may influence blood pressure in developmental and pathological conditions, is poorly understood. Herein, the blood pressure effects of hU-II (0.1-10 nmol/kg) injected intravenously (i.v.) were studied on ketamine/xylazine anesthetized male WKY and SHR rats aged 4 and 8 weeks. hU-II elicited dose-dependent decreases in mean arterial pressure in both strains of animals. The hypotensive responses to hU-II were, however, significantly higher in SHR rats, independently of age. Four-week-old SHR rats (which are normotensive) were, however, less responsive than their hypertensive 8-week-old counterparts. A series of pharmacological inhibitors were used to identify putative endogenous (endothelial) factors that might account for the hU-II-mediated hypotension in 8-week-old SHR. These include the non-selective nitric oxide synthase inhibitor L-NAME (5 micromol/kg), the non-selective cyclooxygenase inhibitor meclofenamate (16 micromol/kg), the voltage-sensitive and ATP-sensitive K+-channel inhibitors, 4-aminopyridine (5 micromol/kg) and glybenclamide (10 micromol/kg), the cytochrome P450 CYP2C9 inhibitor sulfaphenazole (15 micromol/kg), the cytoskeletal fixation agent phalloidin (15 micromol/kg), the endothelin ETB receptor antagonist BQ-788(35 micromol/kg), the bradykinin B2 receptor antagonist HOE 140 (0.5 micromol/kg), the angiotensin AT2 antagonist PD 123319(10 micromol/kg) and the UT receptor antagonist urantide (10 micromol/kg). These agents were administered i.v. either at 2.5, 10 or 40 min prior hU-II injection (10 nmol/kg). Among these inhibitors, sulfaphenazole and phalloidin were able to reduce hU-II-induced hypotension. This suggests that the vasodepressor effect of hU-II is mediated by UT receptors and relies in part on the release of epoxide related products; increased microvascular permeability may also contribute to the blood pressure lowering effect of hU-II. Since urantide blocks the constrictor effects of hU-II on isolated aorta, but is inactive against the hypotensive action of hU-II in vivo, the results presented in this paper provide, for the first time, evidence for the existence of two different functional sites for hU-II.
Collapse
Affiliation(s)
- Gabrielle Gendron
- Department of Pharmacology, Faculty of medicine, Université de Sherbrooke, Sherbrooke, Canada J1H 5N4
| | | | | | | | | | | |
Collapse
|
41
|
Guerrini R, Camarda V, Marzola E, Arduin M, Calo G, Spagnol M, Rizzi A, Salvadori S, Regoli D. Structure-activity relationship study on human urotensin II. J Pept Sci 2005; 11:85-90. [PMID: 15635628 DOI: 10.1002/psc.590] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The vasoactive cyclic undecapeptide urotensin-II (U-II) has been identified as an endogenous ligand for the G-protein coupled receptor now referred to as the UT receptor. The U-II/UT receptor system might be relevant for cardiovascular functions. A structure-activity study of human U-II investigating 31 peptides in the rat aorta bioassay is reported. Ala- and D-scan investigations indicated that the sequence Phe6-Trp7-Lys8-Tyr9 is essential for biological activity and that Lys8 and Tyr9 are particularly important. These two residues were substituted with a series of coded and non-coded amino acids. These studies demonstrated that the positive charge of the primary aliphatic amine at position 8 and its relative spatial orientation is crucial for both receptor occupation and activation, while the only chemical requirement at position 9 is the presence of an aromatic moiety. Moreover, this study led to the identification of UT receptor partial agonists (compounds 23 and 24) which can be used as chemical templates for further investigations aimed at the identification of selective antagonists.
Collapse
Affiliation(s)
- Remo Guerrini
- Department of Pharmaceutical Sciences and Biotechnology Centre, University of Ferrara, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Aiyar N, Johns DG, Ao Z, Disa J, Behm DJ, Foley JJ, Buckley PT, Sarau HM, van-der-Keyl HK, Elshourbagy NA, Douglas SA. Cloning and pharmacological characterization of the cat urotensin-II receptor (UT). Biochem Pharmacol 2005; 69:1069-79. [PMID: 15763543 DOI: 10.1016/j.bcp.2004.12.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Accepted: 12/23/2004] [Indexed: 02/07/2023]
Abstract
Urotensin-II (U-II), acting through its G-protein-coupled receptor, UT, is a possible contributor to hypertension. Variable functional responses to U-II, both within and between species studied to date, complicate the characterization of UT antagonists. In the cat, however, U-II causes systemic hypertension and constricts arterial segments isolated from several vascular beds. The purpose of this study was to clone and pharmacologically characterize cat recombinant UT to determine whether this system represents a model for characterizing UT antagonists. Cloned cat UT displayed 74% identity to primate UT, and 77% identity to rodent UT. [(125)I] hU-II bound in a saturable manner to a single site on recombinant cat UT with high affinity (K(D) 288+/-13pM) and high density (B(max) 747+/-66fmol/mg protein). U-II isopeptides displayed equipotent, high affinity binding to cat UT (K(i) 1.8-5.3nM). Cat UT was coupled to intracellular [Ca(2+)] release (EC(50) 0.6+/-0.2nM) and total inositol phosphate (IP) formation (EC(50) 0.4+/-0.1nM). Protein kinase C activation desensitized cat, but not human, UT-mediated IP formation. UT mRNA expression was detected in cat blood vessels, trachea, lung, and kidney, where the medulla (K(D) 815+/-34) and cortex and (K(D) 316+/-39pM) displayed high affinity binding for human U-II (hU-II). The cat urotensin-II receptor represents a suitable in vitro model to examine the role of the U-II/UT system in the etiology of hypertension, assisting in the evaluation of the UT antagonists to help treat cardiovascular disease.
Collapse
Affiliation(s)
- Nambi Aiyar
- Vascular Biology and Thrombosis Department, Cardiovascular and Urogenital Center for Excellence in Drug Discovery, GlaxoSmithKline, 709 Swedeland Rd, UW2510, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Qi JS, Minor LK, Smith C, Hu B, Yang J, Andrade-Gordon P, Damiano B. Characterization of functional urotensin II receptors in human skeletal muscle myoblasts: comparison with angiotensin II receptors. Peptides 2005; 26:683-90. [PMID: 15752584 DOI: 10.1016/j.peptides.2004.11.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Revised: 11/23/2004] [Accepted: 11/24/2004] [Indexed: 11/22/2022]
Abstract
The properties of urotensin II (U-II) receptor (UT receptor) and angiotensin II (ANG II) receptor (AT receptor) in primary human skeletal myoblasts (HSMM) and differentiated skeletal myotubes (HSMMT) were characterized. Radiolabeled U-II and ANG II bound specifically to HSMM with Kd's of 0.31 nM (2311 receptors/cell) and 0.61 nM (18,257 receptors/cell), respectively. The cyclic segment of U-II peptide, CFWKYC, was the minimal sequence required for binding, with the WKY residues essential. Inhibitor studies suggested AT1 is the predominant ANG II receptor. After radioligand binding, under conditions designed to minimize receptor internalization, half the bound U-II was resistant to acid washing suggesting that U-II binds tightly to its receptor in a quasi-irreversible fashion. The AT1 receptor-bound radioligand was completely removed under the same conditions. RT-PCR detected the expression of mRNAs for UT and AT1 receptors. Western blotting showed that U-II and ANG II signaled via ERK1/2 kinase. UT receptor was not lost upon differentiation into myotubes since both mRNA for UT receptor and U-II binding were still present. ANG II receptors were also present as shown by ANG II-induced calcium mobilization.
Collapse
Affiliation(s)
- Jian-shen Qi
- Vascular Research Team, Johnson and Johnson Pharmaceutical Research and Development, LLC, Welsh and McKean Roads, Spring House, PA 19477-0776, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Urotensin II (UII) has been found to be a potent vasoactive peptide in humans and in a number of relevant animal models of cardiovascular disease such as the mouse, rat and other non-human primates. This peptide with structural homology to somatostatin was first isolated from the urophysis of fish and was recently found to bind to an orphan receptor in mouse and human. Initially found to have potent vasoconstrictive activities in a variety of vessels from diverse species, it has also been shown to exert vasodilatation in certain vessels in the rat and human by various endothelium-dependent mechanisms. The various vasoactive properties of UII suggest that the peptide may have a physiological role in maintaining vascular tone and therefore may have a role in the pathophysiology of a number of human diseases such as heart failure. Moreover, UII has also been implicated as a mitogen of vascular smooth muscle cells suggesting a deleterious role in atherosclerosis and coronary artery disease. In addition, there is evidence to demonstrate that UII has multiple metabolic effects on cholesterol metabolism, glycemic control and hypertension and therefore may be implicated in the development of insulin resistance and the metabolic syndrome.
Collapse
Affiliation(s)
- George Thanassoulis
- Department of Medicine, Montreal General Hospital, McGill University Health Center, 1650 Cedar Avenue, Suite L3-109, Montreal, Quebec H3G 1A4, Canada
| | | | | |
Collapse
|
45
|
Mori M, Fujino M. Urotensin II-related peptide, the endogenous ligand for the urotensin II receptor in the rat brain. Peptides 2004; 25:1815-8. [PMID: 15476951 DOI: 10.1016/j.peptides.2004.06.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 06/02/2004] [Indexed: 11/15/2022]
Abstract
Urotensin II (UII) is a piscine neuropeptide originally isolated from the teleost urophysis. The existence of UII in mammals has been demonstrated by cloning of the mammalian orthologs of UII precursor protein genes. While rat and mouse orthologs have been reported, only the tentative structures of UII peptides of these animals have been demonstrated, since prepro-UII proteins lack the typical processing sites in the amino-terminal region of the mature peptides. A novel peptide, UII-related peptide (URP), was discovered by monitoring UII-immunoreactivity in the rat brain, and its amino acid sequence was determined to be ACFWKYCV. cDNAs encoding rat, mouse, and human precursor proteins for URP were cloned and showed that the sequences of mouse and human URP peptides are identical to that for rat URP. URP was found to bind and activate the human or rat urotensin II receptors [GPR14, UT receptor (UTR)] and showed a hypotensive effect when administrated to anesthetized rats. The prepro-URP gene is expressed in several rat tissues, although with lower levels than the prepro-UII gene and, in the human, is expressed comparably to prepro-UII in several tissues except the spinal cord. These results suggest that URP is the endogenous and functional ligand for urotensin II receptor in the rat and mouse, and possibly in the human.
Collapse
Affiliation(s)
- Masaaki Mori
- Discovery Research Laboratories, Pharmaceutical Research Division, Takeda Chemical Industries Ltd., Wadai 10, Tsukuba, Ibaraki 300-4293, Japan.
| | | |
Collapse
|
46
|
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: 1.6] [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
|
47
|
Onan D, Pipolo L, Yang E, Hannan RD, Thomas WG. Urotensin II Promotes Hypertrophy of Cardiac Myocytes via Mitogen-Activated Protein Kinases. Mol Endocrinol 2004; 18:2344-54. [PMID: 15205471 DOI: 10.1210/me.2003-0309] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Urotensin II and its receptor are coexpressed in the heart and up-regulated during cardiac dysfunction. In cultured neonatal cardiomyocytes, we mimicked this up-regulation using an adenovirus to increase expression of the urotensin receptor. In this model system, urotensin II promoted strong hypertrophic growth and phenotypic changes, including cell enlargement and sarcomere reorganization. Urotensin II potently activated the MAPKs, ERK1/2 and p38, and blocking these kinases with PD098059 and SB230580, respectively, significantly inhibited urotensin II-mediated hypertrophy. In contrast, urotensin II did not activate JNK. The activation of ERK1/2 and p38 as well as cellular hypertrophy was independent of protein kinase C, and calcium and phosphoinositide 3-kinase, yet dependent on the capacity of the urotensin receptor to trans-activate the epidermal growth factor receptor. Urotensin II promoted the tyrosine phosphorylation of epidermal growth factor receptors, which was inhibited by the selective epidermal growth factor receptor kinase inhibitor, AG1478. These data indicate that perturbations in cardiac homeostasis, which lead to up-regulation of urotensin II receptors, promote urotensin II-mediated cardiomyocyte hypertrophy via ERK1/2 and p38 signaling pathways in an epidermal growth factor receptor-dependent manner.
Collapse
Affiliation(s)
- Döne Onan
- Molecular Endocrinology, Baker Heart Research Institute, P.O. Box 6492, St. Kilda Road Central, Melbourne 8008, Victoria, Australia
| | | | | | | | | |
Collapse
|
48
|
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
|
49
|
Aiyar N, Guida B, Ao Z, Disa J, Naselsky D, Behm DJ, Su JL, Kull FC, Douglas SA. Differential levels of "urotensin-II-like" activity determined by radio-receptor and radioimmuno-assays. Peptides 2004; 25:1339-47. [PMID: 15350702 DOI: 10.1016/j.peptides.2004.05.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Revised: 05/20/2004] [Accepted: 05/20/2004] [Indexed: 02/07/2023]
Abstract
Plasma and urinary levels of "urotensin(U)-II-like" substances determined in healthy human volunteers were 12.4 +/- 0.6 ng/ml and 2.2 +/- 0.3 ng/ml by RIA, an order of magnitude lower than that seen by RRA, 167.5 +/- 9.5 ng/ml and 65.2 +/- 4.3 ng/ml. HPLC demonstrated the existence of at least three prominent activity peaks in plasma and urine, the more hydrophobic of which did not co-elute with U-II, degradation products or URP. RRA and RIA recognized these peaks with contrasting efficacy. As such, published levels of "U-II-like" activity should be interpreted with caution until a better understanding is obtained regarding what species specific RIA and RRA assay reagents interact with.
Collapse
Affiliation(s)
- Nambi Aiyar
- Department of Vascular Biology and Thrombosis (UW2510), Cardiovascular and Urogenital Center of Excellence for Drug Discovery (CEDD), GlaxoSmithKline, P.O. Box 1539, 709 Swedeland Road, King of Prussia, PA 19406-0939, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Douglas SA, Naselsky D, Ao Z, Disa J, Herold CL, Lynch F, Aiyar NV. Identification and pharmacological characterization of native, functional human urotensin-II receptors in rhabdomyosarcoma cell lines. Br J Pharmacol 2004; 142:921-32. [PMID: 15210573 PMCID: PMC1575108 DOI: 10.1038/sj.bjp.0705743] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1 In an effort to identify endogenous, native mammalian urotensin-II (U-II) receptors (UT), a diverse range of human, primate and rodent cell lines (49 in total) were screened for the presence of detectable [125I]hU-II binding sites. 2 UT mRNA (Northern blot, PCR) and protein (immunocytochemistry) were evident in human skeletal muscle tissue and cells. 3 [(125)I]hU-II bound to a homogenous population of high-affinity, saturable (Kd 67.0+/-11.8 pm, Bmax 9687+/-843 sites cell(-1)) receptors in the skeletal muscle (rhabdomyosarcoma) cell line SJRH30. Radiolabel was characteristically slow to dissociate (< or =15% dissociation 90 min). A lower density of high-affinity U-II binding sites was also evident in the rhabdomyosarcoma cell line TE671 (1667+/-165 sites cell(-1), Kd 74+/-8 pm). 4 Consistent with the profile recorded in human recombinant UT-HEK293 cells, [125I]hU-II binding to SJRH30 cells was selectively displaced by both mammalian and fish U-II isopeptides (Kis 0.5+/-0.1-1.2+/-0.3 nm) and related analogues (hU-II[4-11]>[Cys(5,10)]Acm hU-II; Kis 0.4+/-0.1 and 864+/-193 nm, respectively). 5 U-II receptor activation was functionally coupled to phospholipase C-mediated [Ca2+]i mobilization (EC50 6.9+/-2.2 nm) in SJRH30 cells. 6 The present study is the first to identify the presence of 'endogenous' U-II receptors in SJRH30 and TE671 cells. SJRH30 cells, in particular, might prove to be of utility for (a) investigating the pharmacological properties of hU-II and related small molecule antagonists at native human UT and (b) delineating the role of this neuropeptide in the (patho)physiological regulation of mammalian neuromuscular function.
Collapse
Affiliation(s)
- Stephen A Douglas
- Department of Vascular Biology and Thrombosis, Cardiovascular and Urogenital Center of Excellence for Drug Discovery, GlaxoSmithKline, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | |
Collapse
|