1
|
Nassour H, Pétrin D, Devost D, Billard E, Sleno R, Hébert TE, Chatenet D. Evidence for heterodimerization and functional interaction of the urotensin II and the angiotensin II type 1 receptors. Cell Signal 2024; 116:111056. [PMID: 38262555 DOI: 10.1016/j.cellsig.2024.111056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
Despite the observation of synergistic interactions between the urotensinergic and angiotensinergic systems, the interplay between the urotensin II receptor (hUT) and the angiotensin II type 1 receptor (hAT1R) in regulating cellular signaling remains incompletely understood. Notably, the putative interaction between hUT and hAT1R could engender reciprocal allosteric modulation of their signaling signatures, defining a unique role for these complexes in cardiovascular physiology and pathophysiology. Using a combination of co-immunoprecipitation, bioluminescence resonance energy transfer (BRET) and FlAsH BRET-based conformational biosensors, we first demonstrated the physical interaction between hUT and hAT1R. Next, to analyze how this functional interaction regulated proximal and distal hUT- and hAT1R-associated signaling pathways, we used BRET-based signaling biosensors and western blots to profile pathway-specific signaling in HEK 293 cells expressing hUT, hAT1R or both. We observed that hUT-hAT1R heterodimers triggered distinct signaling outcomes compared to their respective parent receptors alone. Notably, co-transfection of hUT and hAT1R has no impact on hUII-induced Gq activation but significantly reduced the potency and efficacy of Ang II to mediate Gq activation. Interestingly, URP, the second hUT endogenous ligand, produce a distinct signaling signature compared to hUII at hUT-hAT1R. Our results therefore suggest that assembly of hUT with hAT1R might be important for allosteric modulation of outcomes associated with specific hardwired signaling complexes in healthy and disease states. Altogether, our work, which potentially explains the interplay observed in native cells and tissues, validates such complexes as potential targets to promote the design of compounds that can modulate heterodimer function selectively.
Collapse
Affiliation(s)
- Hassan Nassour
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Ville de Laval, QC, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Etienne Billard
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Rory Sleno
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada.
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Ville de Laval, QC, Canada.
| |
Collapse
|
2
|
Rex DAB, Suchitha GP, Palollathil A, Kanichery A, Prasad TSK, Dagamajalu S. The network map of urotensin-II mediated signaling pathway in physiological and pathological conditions. J Cell Commun Signal 2022; 16:601-608. [PMID: 35174439 PMCID: PMC9733756 DOI: 10.1007/s12079-022-00672-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Urotensin-II is a polypeptide ligand with neurohormone-like activity. It mediates downstream signaling pathways through G-protein-coupled receptor 14 (GPR14) also known as urotensin receptor (UTR). Urotensin-II is the most potent endogenous vasoconstrictor in mammals, promoting cardiovascular remodelling, cardiac fibrosis, and cardiomyocyte hypertrophy. It is also involved in other physiological and pathological activities, including neurosecretory effects, insulin resistance, atherosclerosis, kidney disease, and carcinogenic effects. Moreover, it is a notable player in the process of inflammatory injury, which leads to the development of inflammatory diseases. Urotensin-II/UTR expression stimulates the accumulation of monocytes and macrophages, which promote the adhesion molecules expression, chemokines activation and release of inflammatory cytokines at inflammatory injury sites. Therefore, urotensin-II turns out to be an important therapeutic target for the treatment options and management of associated diseases. The main downstream signaling pathways mediated through this urotensin-II /UTR system are RhoA/ROCK, MAPKs and PI3K/AKT. Due to the importance of urotensin-II systems in biomedicine, we consolidated a network map of urotensin-II /UTR signaling. The described signaling map comprises 33 activation/inhibition events, 31 catalysis events, 15 molecular associations, 40 gene regulation events, 60 types of protein expression, and 11 protein translocation events. The urotensin-II signaling pathway map is made freely accessible through the WikiPathways Database ( https://www.wikipathways.org/index.php/Pathway:WP5158 ). The availability of comprehensive urotensin-II signaling in the public resource will help understand the regulation and function of this pathway in normal and pathological conditions. We believe this resource will provide a platform to the scientific community in facilitating the identification of novel therapeutic drug targets for diseases associated with urotensin-II signaling.
Collapse
Affiliation(s)
- D. A. B. Rex
- grid.413027.30000 0004 1767 7704Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - G. P. Suchitha
- grid.413027.30000 0004 1767 7704Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - Akhina Palollathil
- grid.413027.30000 0004 1767 7704Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - Anagha Kanichery
- grid.413027.30000 0004 1767 7704Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - T. S. Keshava Prasad
- grid.413027.30000 0004 1767 7704Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| | - Shobha Dagamajalu
- grid.413027.30000 0004 1767 7704Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575018 India
| |
Collapse
|
3
|
Chen L, Yu D, Ling S, Xu JW. Mechanism of tonifying-kidney Chinese herbal medicine in the treatment of chronic heart failure. Front Cardiovasc Med 2022; 9:988360. [PMID: 36172573 PMCID: PMC9510640 DOI: 10.3389/fcvm.2022.988360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 12/04/2022] Open
Abstract
According to traditional Chinese medicine (TCM), chronic heart failure has the basic pathological characteristics of “heart-kidney yang deficiency.” Chronic heart failure with heart- and kidney-Yang deficiency has good overlap with New York Heart Association (NYHA) classes III and IV. Traditional Chinese medicine classical prescriptions for the treatment of chronic heart failure often take “warming and tonifying kidney-Yang” as the core, supplemented by herbal compositions with functions of “promoting blood circulation and dispersing blood stasis.” Nowadays, there are still many classical and folk prescriptions for chronic heart failure treatment, such as Zhenwu decoction, Bushen Huoxue decoction, Shenfu decoction, Sini decoction, as well as Qili Qiangxin capsule. This review focuses on classical formulations and their active constituents that play a key role in preventing chronic heart failure by suppressing inflammation and modulating immune and neurohumoral factors. In addition, given that mitochondrial metabolic reprogramming has intimate relation with inflammation, cardiac hypertrophy, and fibrosis, the regulatory role of classical prescriptions and their active components in metabolic reprogramming, including glycolysis and lipid β-oxidation, is also presented. Although the exact mechanism is unknown, the classical TCM prescriptions still have good clinical effects in treating chronic heart failure. This review will provide a modern pharmacological explanation for its mechanism and offer evidence for clinical medication by combining TCM syndrome differentiation with chronic heart failure clinical stages.
Collapse
|
4
|
Mazurara GR, Dallagnol JCC, Chatenet D, Allen BG, Hébert TE. The complicated lives of GPCRs in cardiac fibroblasts. Am J Physiol Cell Physiol 2022; 323:C813-C822. [PMID: 35938678 DOI: 10.1152/ajpcell.00120.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of different G protein-coupled receptors (GPCRs) in the cardiovascular system is well understood in cardiomyocytes and vascular smooth muscle cells (VSMCs). In the former, stimulation of Gs-coupled receptors leads to increases in contractility, while stimulation of Gq-coupled receptors modulates cellular survival and hypertrophic responses. In VSMCs, stimulation of GPCRs also modulates contractile and cell growth phenotypes. Here, we will focus on the relatively less well studied effects of GPCRs in cardiac fibroblasts, focusing on key signalling events involved in the activation and differentiation of these cells. We also review the hierarchy of signalling events driving the fibrotic response and the communications between fibroblasts and other cells in the heart. We discuss how such events may be distinct depending on where the GPCRs and their associated signalling machinery are localized in these cells with an emphasis on nuclear membrane-localized receptors. Finally, we explore what such connections between cell surface and nuclear GPCR signalling might mean for cardiac fibrosis.
Collapse
Affiliation(s)
- Grace R Mazurara
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | - Juliana C C Dallagnol
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada.,Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Laval, Québec, Canada.,Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Laval, Québec, Canada
| | - Bruce G Allen
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| |
Collapse
|
5
|
Bourque K, Hawey C, Jones-Tabah J, Pétrin D, Martin RD, Ling Sun Y, Hébert TE. Measuring hypertrophy in neonatal rat primary cardiomyocytes and human iPSC-derived cardiomyocytes. Methods 2021; 203:447-464. [PMID: 34933120 DOI: 10.1016/j.ymeth.2021.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
In the heart, left ventricular hypertrophy is initially an adaptive mechanism that increases wall thickness to preserve normal cardiac output and function in the face of coronary artery disease or hypertension. Cardiac hypertrophy develops in response to pressure and volume overload but can also be seen in inherited cardiomyopathies. As the wall thickens, it becomes stiffer impairing the distribution of oxygenated blood to the rest of the body. With complex cellular signalling and transcriptional networks involved in the establishment of the hypertrophic state, several model systems have been developed to better understand the molecular drivers of disease. Immortalized cardiomyocyte cell lines, primary rodent and larger animal models have all helped understand the pathological mechanisms underlying cardiac hypertrophy. Induced pluripotent stem cell-derived cardiomyocytes are also used and have the additional benefit of providing access to human samples with direct disease relevance as when generated from patients suffering from hypertrophic cardiomyopathies. Here, we briefly review in vitro and in vivo model systems that have been used to model hypertrophy and provide detailed methods to isolate primary neonatal rat cardiomyocytes as well as to generate cardiomyocytes from human iPSCs. We also describe how to model hypertrophy in a "dish" using gene expression analysis and immunofluorescence combined with automated high-content imaging.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Yi Ling Sun
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
6
|
Schuster R, Steffen P, Dreyer B, Rohn S, Schlüter H, Riedner M. Identifying Circulating Urotensin II and Urotensin II-Related Peptide-Generating Enzymes in the Human Plasma Fraction Cohn IV-4. J Proteome Res 2021; 20:5368-5378. [PMID: 34734734 DOI: 10.1021/acs.jproteome.1c00521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Urotensin II (UII) and UII-related peptide (URP) are vasoactive peptide hormones causing strong vasoconstriction or vasodilation, depending on the type of blood vessel. In humans, the active forms are resulting from proteolytic cleavage of their inactive precursor protein. In blood plasma, a defined protease converting the inactive UII and URP precursors into their active forms has not been identified yet. Using mass spectrometry-based enzyme screening for detecting UII- and URP-converting enzymes, the human plasma fraction Cohn IV-4 was chromatographed, and the resulting fractions were screened for UII- or URP-generating activity. Plasma kallikrein (PK) as a UII- and URP-generating protease was identified. URP generation was also found for the serine protease factor XIa, plasmin, thrombin, and, to a smaller extent, factor XIIa. It was demonstrated that in the Cohn IV-4 fraction, PK accounts for a significant amount of UII- and URP-generating activity.
Collapse
Affiliation(s)
- Raphael Schuster
- Institute of Organic Chemistry, Department of Chemistry, Universität Hamburg, 20146 Hamburg, Germany
| | - Pascal Steffen
- Bowel Cancer & Biomarker Lab, Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, New South Wales 2065, Australia
| | - Benjamin Dreyer
- Mass Spectrometric Proteomics, Institute of Clinical Chemistry, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sascha Rohn
- Hamburg School of Food Science, Institute of Food Chemistry, Universität Hamburg, Grindelallee 117, 20146 Hamburg, Germany.,Institute of Food Technology and Food Chemistry, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Hartmut Schlüter
- Mass Spectrometric Proteomics, Institute of Clinical Chemistry, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Riedner
- Institute of Organic Chemistry, Department of Chemistry, Universität Hamburg, 20146 Hamburg, Germany
| |
Collapse
|
7
|
Yu QQ, Cheng DX, Xu LR, Li YK, Zheng XY, Liu Y, Li YF, Liu HL, Bai L, Wang R, Fan JL, Liu EQ, Zhao SH. Urotensin II and urantide exert opposite effects on the cellular components of atherosclerotic plaque in hypercholesterolemic rabbits. Acta Pharmacol Sin 2020; 41:546-553. [PMID: 31685976 PMCID: PMC7468446 DOI: 10.1038/s41401-019-0315-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/30/2019] [Indexed: 12/26/2022]
Abstract
Increasing levels of plasma urotensin II (UII) are positively associated with atherosclerosis. In this study we investigated the role of macrophage-secreted UII in atherosclerosis progression, and evaluated the therapeutic value of urantide, a potent competitive UII receptor antagonist, in atherosclerosis treatment. Macrophage-specific human UII-transgenic rabbits and their nontransgenic littermates were fed a high cholesterol diet for 16 weeks to induce atherosclerosis. Immunohistochemical staining of the cellular components (macrophages and smooth muscle cells) of aortic atherosclerotic lesions revealed a significant increase (52%) in the macrophage-positive area in only male transgenic rabbits compared with that in the nontransgenic littermates. However, both male and female transgenic rabbits showed a significant decrease (45% in males and 31% in females) in the smooth muscle cell-positive area compared with that of their control littermates. The effects of macrophage-secreted UII on the plaque cellular components were independent of plasma lipid level. Meanwhile the wild-type rabbits were continuously subcutaneously infused with urantide (5.4 µg· kg-1· h-1) using osmotic mini-pumps. Infusion of urantide exerted effects opposite to those caused by UII, as it significantly decreased the macrophage-positive area in male wild-type rabbits compared with that of control rabbits. In cultured human umbilical vein endothelial cells, treatment with UII dose-dependently increased the expression of the adhesion molecules VCAM-1 and ICAM-1, and this effect was partially reversed by urantide. The current study provides direct evidence that macrophage-secreted UII plays a key role in atherogenesis. Targeting UII with urantide may promote plaque stability by decreasing macrophage-derived foam cell formation, which is an indicator of unstable plaque.
Collapse
|
8
|
Oral E, Halici Z, Cinar I, Ozcan E, Kutlu Z. Evaluation of Endothelial Dysfunction in Bipolar Affective Disorders: Serum Endocan and Urotensin-II Levels. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2019; 17:211-221. [PMID: 30905121 PMCID: PMC6478082 DOI: 10.9758/cpn.2019.17.2.211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/24/2018] [Accepted: 04/22/2018] [Indexed: 12/11/2022]
Abstract
Objective This study investigated changes in urotensin-II (U-II) and endocan levels which can be used as an early biological marker of endothelial injury in the episode and remission phases of bipolar affective disorder (BAD). Methods We compared endocan and U-II levels, which has been shown to be closely associated with neurotransmitter systems in addition to continuity of endothelial structure and inflammatory response, in patients with BAD in remission for at least one year (n=42) and in patients still in manic or depressive episodes (n=16) with healthy controls (n=30). Results Both endocan and U-II levels were significantly higher in the bipolar patients than in the controls. Endocan and U-II levels were also significantly correlated with one another (p=0.000, r=0.833). Both endocan (p=0.000) and U-II levels (p=0.000) were significantly higher in the bipolar attack group compared to the subjects in remission, and in the remission group compared to the controls. Conclusion In this study we determined significantly higher endocan and U-II levels in BAD compared to the controls, while serum endocan and U-II levels of patients undergoing attacks were also significantly higher than those of the controls and also those of patients in remission.
Collapse
Affiliation(s)
- Elif Oral
- Department of Psychiatry, Faculty of Medicine, Izmir Katip Çelebi University
| | - Zekai Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University
| | - Irfan Cinar
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University
| | - Elif Ozcan
- Department of Psychiatry, Erzurum Regional Education and Research Hospital
| | - Zerrin Kutlu
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University
| |
Collapse
|
9
|
Lim CJ, Kim NH, Park HJ, Lee BH, Oh KS, Yi KY. Synthesis and SAR of 5-aryl-furan-2-carboxamide derivatives as potent urotensin-II receptor antagonists. Bioorg Med Chem Lett 2019; 29:577-580. [PMID: 30611618 DOI: 10.1016/j.bmcl.2018.12.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/20/2018] [Accepted: 12/26/2018] [Indexed: 01/27/2023]
Abstract
The synthesis and biological evaluation as potential urotensin-II receptor antagonists of a series of 5-arylfuran-2-carboxamide derivatives 1, bearing a 4-(3-chloro-4-(piperidin-4-yloxy)benzyl)piperazin-1-yl group, are described. The results of a systematic SAR investigation of furan-2-carboxamides with C-5 aryl groups possessing a variety of aryl ring substituents led to identification of the 3,4-difluorophenyl analog 1y as a highly potent UT antagonist with an IC50 value of 6 nM. In addition, this substance was found to display high metabolic stability, and low hERG inhibition and cytotoxicity, and to have an acceptable PK profile.
Collapse
Affiliation(s)
- Chae Jo Lim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, KRICT School, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea.
| | - Nam Hui Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, KRICT School, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hye Jin Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, KRICT School, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Byung Ho Lee
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Kwang-Seok Oh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, KRICT School, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Kyu Yang Yi
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Yuseong-gu, Daejeon 34114, Republic of Korea; Department of Medicinal Chemistry and Pharmacology, KRICT School, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea.
| |
Collapse
|
10
|
Sun SL, Liu LM. Urotensin II: an inflammatory cytokine. J Endocrinol 2019; 240:JOE-18-0505.R2. [PMID: 30601760 DOI: 10.1530/joe-18-0505] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022]
Abstract
Urotensin II (UII) is a polypeptide molecule with neurohormone-like activity. It has been confirmed that UII is widely distributed in numerous organs of different animal species from fish to mammals, including humans. The UII receptor is orphan G-protein coupled receptor 14, also known as UT. The tissue distribution of UII and UT is highly consistent, and their expression may be regulated by autocrine and paracrine mechanisms. In the body, UII has many physiological and pathophysiological activities, such as vasoconstrictor and vasodilatory actions, cell proliferation, pro-fibrosis, neuroendocrine activity, insulin resistance, and carcinogenic and inflammatory effects, which have been recognized only in recent years. In fact, UII is involved in the process of inflammatory injury and plays a key role in the onset and development of inflammatory diseases. In this paper, we will review the roles UII plays in inflammatory diseases.
Collapse
Affiliation(s)
- Sui-Lin Sun
- S Sun, Department of Infection, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China, Nanchang, China
| | - Liang-Ming Liu
- L Liu, Department of Infection, Songjiang Hospital Affiliated to First People's Hospital, Shanghai Jiaotong University, Shanghai, 201600, China
| |
Collapse
|
11
|
Trotta MC, Maisto R, Alessio N, Hermenean A, D'Amico M, Di Filippo C. The Melanocortin MC5R as a New Target for Treatment of High Glucose-Induced Hypertrophy of the Cardiac H9c2 Cells. Front Physiol 2018; 9:1475. [PMID: 30416452 PMCID: PMC6212602 DOI: 10.3389/fphys.2018.01475] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022] Open
Abstract
The study explored the anti-hypertrophic effect of the melanocortin MC5R stimulation in H9c2 cardiac myocytes exposed to high glucose. This has been done by using α-MSH and selective MC5R agonists and assessing the expression of GLUT4 and GLUT1 transporters, miR-133 and urotensin receptor levels as a marker of cardiac hypertrophy. The study shows for the first time an up-regulation of MC5R expression levels in H9c2 cardiomyocytes exposed to high glucose medium (33 mM D-glucose) for 48 h, compared to cells grown in normal glucose medium (5.5 mM D-glucose). Moreover, H9c2 cells exposed to high glucose showed a significant reduction in cell viability (-40%), a significant increase in total protein per cell number (+109%), and an increase of the urotensin receptor expression levels as an evidence of cells hypertrophy. The pharmacological stimulation of MC5R with α-MSH (90 pM)of the high glucose exposed H9c2 cells increased the cell survival (+50,8%) and reduced the total protein per cell number (-28,2%) with respect to high glucose alone, confirming a reduction of the hypertrophic state as per cell area measurement. Similarly, PG-901 (selective agonist, 10-10 M) significantly increased cell viability (+61,0 %) and reduced total protein per cell number (-40,2%), compared to cells exposed to high glucose alone. Interestingly, the MC5R agonist reduced the GLUT1/GLUT4 glucose transporters ratio on the cell membranes exhibited by the hypertrophic H9c2 cells and increased the intracellular PI3K activity, mediated by a decrease of the levels of the miRNA miR-133a. The beneficial effects of MC5R agonism on the cardiac hypertrophy caused by high glucose was also observed also by echocardiographic evaluations of rats made diabetics with streptozotocin (65 mg/kg i.p.). Therefore, the melanocortin MC5R could be a new target for the treatment of high glucose-induced hypertrophy of the cardiac H9c2 cells.
Collapse
Affiliation(s)
- Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Anca Hermenean
- Institute of Life Sciences, "Vasile Goldis" Western University of Arad, Arad, Romania
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Clara Di Filippo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
12
|
Urotensin receptors as a new target for CLP induced septic lung injury in mice. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:135-145. [DOI: 10.1007/s00210-018-1571-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/16/2018] [Indexed: 12/27/2022]
|
13
|
OKUYAN HAMZAMALİK, TERZİ MENDERESYUSUF, ÖNLEN GÜNERİ CANSU, URHAN KÜÇÜK MERAL. Effect of Pro-Inflammatory Cytokine IL-1β, on Urotensin II Gene Expression in Human Lung Cancer Cells. KONURALP TIP DERGISI 2018. [DOI: 10.18521/ktd.398669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
14
|
Sung JH, Song A, Park T, Kim E, Lee S. The Different Expression Patterns of HSP22, a Late Embryogenesis Abundant-like Protein, in Hypertrophic H9C2 Cells Induced by NaCl and Angiotensin II. Electrolyte Blood Press 2018; 16:1-10. [PMID: 30046328 PMCID: PMC6051945 DOI: 10.5049/ebp.2018.16.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/25/2018] [Indexed: 11/05/2022] Open
Abstract
Background High-NaCl diet is a contributing factor for cardiac hypertrophy. The role of HSP22 as a protective protein during cardiac hypertrophy due to hypernatremia is unclear. Accordingly, this study aimed to establish a cellular hypernatremic H9C2 model and to compare the expression of HSP22 in Ca2+ homeostasis between a high-NaCl and angiotensin II-induced hypertrophic cellular H9C2 model. Methods Real-time PCR was performed to compare the mRNA expression. Flow cytometry and confocal microscopy were used to analyze the cells. Results The addition of 30 mM NaCl for 48 h was the most effective condition for the induction of hypertrophic H9C2 cells (termed the in vitro hypernatremic model). Cardiac cellular hypertrophy was induced with 30 mM NaCl and 1 µM angiotensin II for 48 h, without causing abnormal morphological changes or cytotoxicity of the culture conditions. HSP22 contains a similar domain to that found in the consensus sequences of the late embryogenesis abundant protein group 3 from Artemia. The expression of HSP22 gradually decreased in the in vitro hypernatremic model. In contrast to the in vitro hypernatremic model, HSP22 increased after exposure to angiotensin II for 48 h. Intracellular Ca2+ decreased in the angiotensin II model and further decreased in the in vitro hypernatremic model. Impaired intracellular Ca2+ homeostasis was more evident in the in vitro hypernatremic model. Conclusion The results showed that NaCl significantly decreased HSP22. Decreased HSP22, due to the hypernatremic condition, affected the Ca2+ homeostasis in the H9C2 cells. Therefore, hypernatremia induces cellular hypertrophy via impaired Ca2+ homeostasis. The additional mechanisms of HSP22 need to be explored further.
Collapse
Affiliation(s)
- Jae Hwi Sung
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Ahran Song
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Taegun Park
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Eunyoung Kim
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| | - Seunggwan Lee
- Integrated Biomedical and Life Science, College of Health Science, Korea University, Seoul, Korea
| |
Collapse
|
15
|
Jumaah S, Çelekli A, Sucu M. The role of human urotensin-II in patients with hypertrophic cardiomyopathy. J Immunoassay Immunochem 2018; 39:150-162. [PMID: 28686108 DOI: 10.1080/15321819.2017.1344130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Hypertrophic cardiomyopathy (HCM) is a genetic condition with the hallmark feature of left ventricular hypertrophy. Human Urotensin-II (hUT-II) is regarded as a cardiovascular autacoid/hormone, and it has cardiac inotropic and hypertrophic properties. Aims of this study were to elucidate the clinical significance of serum hUT-II levels as a potential new biomarker in patients with HCM. METHODS This study included 40 HCM patients (60% males and 40% females) and were compared to 30 healthy control subjects (47% males and 53% females. All patients underwent extensive clinical, laboratory, and echocardiographic. Blood samples were taken to test for serum hUT-II levels by commercial ELISA Kit. RESULTS Serum hUT-II was significantly higher (p < 0.01) in patients with HCM (15.8 ± 2.1 pmol/L) compared with healthy controls (3.3 ± 1.7 pmol/L). With regard to HCM patient, Serum hUT-II levels were significantly higher in the female with 16.3 ± 1.9 pmol/L than the male with 15.4 ± 2.2 pmol/L (p < 0.05). Among echocardiographic parameters, hUT-II was negatively associated with ejection fraction (r = -0.160, p = 0.324). CONCLUSION Results of the first study indicated that serum hUT-II levels were markedly elevated in patients with HCM. Serum hUT-II is a novel biomarker parameter that has clinical use in patients with the severity of LVH.
Collapse
Affiliation(s)
- Saman Jumaah
- a Institute of Natural and Applied Sciences, Department of Biochemistry Science and Technology , Gaziantep University , Gaziantep , Turkey
| | - Abuzer Çelekli
- b Faculty of Arts and Science, Department of Biology , Gaziantep University , Gaziantep , Turkey
| | - Murat Sucu
- c Medical Faculty Cardiology , Gaziantep University , Gaziantep , Turkey
| |
Collapse
|
16
|
Billard É, Iddir M, Nassour H, Lee-Gosselin L, Poujol de Molliens M, Chatenet D. New directions for urotensin II receptor ligands. Pept Sci (Hoboken) 2018. [DOI: 10.1002/pep2.24056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Étienne Billard
- INRS-Institut Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec; Ville de Laval Québec H7V 1B7 Canada
| | - Mustapha Iddir
- INRS-Institut Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec; Ville de Laval Québec H7V 1B7 Canada
| | - Hassan Nassour
- INRS-Institut Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec; Ville de Laval Québec H7V 1B7 Canada
| | - Laura Lee-Gosselin
- INRS-Institut Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec; Ville de Laval Québec H7V 1B7 Canada
| | - Mathilde Poujol de Molliens
- INRS-Institut Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec; Ville de Laval Québec H7V 1B7 Canada
| | - David Chatenet
- INRS-Institut Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec; Ville de Laval Québec H7V 1B7 Canada
| |
Collapse
|
17
|
Li J, Zhao PP, Hao T, Wang D, Wang Y, Zhu YZ, Wu YQ, Zhou CH. Urotensin II inhibitor eases neuropathic pain by suppressing the JNK/NF-κB pathway. J Endocrinol 2017; 232:165-174. [PMID: 27895138 DOI: 10.1530/joe-16-0255] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022]
Abstract
Urotensin II (U-II), a cyclic peptide originally isolated from the caudal neurosecretory system of fishes, can produce proinflammatory effects through its specific G protein-coupled receptor, GPR14. Neuropathic pain, a devastating disease, is related to excessive inflammation in the spinal dorsal horn. However, the relationship between U-II and neuropathic pain has not been reported. This study was designed to investigate the effect of U-II antagonist on neuropathic pain and to understand the associated mechanisms. We reported that U-II and its receptor GPR14 were persistently upregulated and activated in the dorsal horn of L4-6 spinal cord segments after chronic constriction injury (CCI) in rats. Intrathecal injection of SB657510, a specific antagonist against U-II, reversed CCI-induced thermal hyperalgesia and mechanical allodynia. Furthermore, we found that SB657510 reduced the expression of phosphorylated c-Jun N-terminal kinase (p-JNK) and nuclear factor-κB (NF-κB) p65 as well as subsequent secretion of interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α). It was also showed that both the JNK inhibitor SP600125 and the NF-κB inhibitor PDTC significantly attenuated thermal hyperalgesia and mechanical allodynia in CCI rats. Our present research showed that U-II receptor antagonist alleviated neuropathic pain possibly through the suppression of the JNK/NF-κB pathway in CCI rats, which will contribute to the better understanding of function of U-II and pathogenesis of neuropathic pain.
Collapse
Affiliation(s)
- Jing Li
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
| | - Pan-Pan Zhao
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
| | - Ting Hao
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
| | - Dan Wang
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
| | - Yu Wang
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
| | - Yang-Zi Zhu
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
- Department of Anesthetic PharmacologyXuzhou Medical University, Xuzhou, China
| | - Cheng-Hua Zhou
- Jiangsu Province Key Laboratory of AnesthesiologyXuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical PharmacyXuzhou Medical University, Xuzhou, China
| |
Collapse
|
18
|
Castel H, Desrues L, Joubert JE, Tonon MC, Prézeau L, Chabbert M, Morin F, Gandolfo P. The G Protein-Coupled Receptor UT of the Neuropeptide Urotensin II Displays Structural and Functional Chemokine Features. Front Endocrinol (Lausanne) 2017; 8:76. [PMID: 28487672 PMCID: PMC5403833 DOI: 10.3389/fendo.2017.00076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/28/2017] [Indexed: 12/16/2022] Open
Abstract
The urotensinergic system was previously considered as being linked to numerous physiopathological states, including atherosclerosis, heart failure, hypertension, pre-eclampsia, diabetes, renal disease, as well as brain vascular lesions. Thus, it turns out that the actions of the urotensin II (UII)/G protein-coupled receptor UT system in animal models are currently not predictive enough in regard to their effects in human clinical trials and that UII analogs, established to target UT, were not as beneficial as expected in pathological situations. Thus, many questions remain regarding the overall signaling profiles of UT leading to complex involvement in cardiovascular and inflammatory responses as well as cancer. We address the potential UT chemotactic structural and functional definition under an evolutionary angle, by the existence of a common conserved structural feature among chemokine receptorsopioïdergic receptors and UT, i.e., a specific proline position in the transmembrane domain-2 TM2 (P2.58) likely responsible for a kink helical structure that would play a key role in chemokine functions. Even if the last decade was devoted to the elucidation of the cardiovascular control by the urotensinergic system, we also attempt here to discuss the role of UII on inflammation and migration, likely providing a peptide chemokine status for UII. Indeed, our recent work established that activation of UT by a gradient concentration of UII recruits Gαi/o and Gα13 couplings in a spatiotemporal way, controlling key signaling events leading to chemotaxis. We think that this new vision of the urotensinergic system should help considering UT as a chemotactic therapeutic target in pathological situations involving cell chemoattraction.
Collapse
Affiliation(s)
- Hélène Castel
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- *Correspondence: Hélène Castel,
| | - Laurence Desrues
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Jane-Eileen Joubert
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Marie-Christine Tonon
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Laurent Prézeau
- CNRS UMR 5203, INSERM U661, Institute of Functional Genomic (IGF), University of Montpellier 1 and 2, Montpellier, France
| | - Marie Chabbert
- UMR CNRS 6214, INSERM 1083, Faculté de Médecine 3, Angers, France
| | - Fabrice Morin
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Pierrick Gandolfo
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
19
|
Zhao J, Jiang J, Wang J, Liu L, Han XN, Chu SY, Xue L, Ding WH. Genetic polymorphisms ofUTS2rs2890565 Ser89Asn in cardiac hypertrophy in Chinese Han population. Postgrad Med J 2016; 93:406-413. [DOI: 10.1136/postgradmedj-2016-134476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/25/2016] [Accepted: 11/15/2016] [Indexed: 01/12/2023]
|
20
|
Benzo[ b ]thiophene-2-carboxamide derivatives as potent urotensin-II receptor antagonists. Bioorg Med Chem Lett 2016; 26:4684-4686. [DOI: 10.1016/j.bmcl.2016.08.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/12/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023]
|
21
|
Liu LM, Tu WJ, Zhu T, Wang XT, Tan ZL, Zhong H, Gao DY, Liang DY. IRF3 is an important molecule in the UII/UT system and mediates immune inflammatory injury in acute liver failure. Oncotarget 2016; 7:49027-49041. [PMID: 27448985 PMCID: PMC5226488 DOI: 10.18632/oncotarget.10717] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022] Open
Abstract
The urotensin II/urotensin receptor (UII/UT) system can mediate inflammatory liver injury in acute liver failure (ALF); however; the related mechanism is not clear. In this study, we confirmed that lipopolysaccharide/D-galactosamine (LPS/D-GalN) induced up-regulation of liver interferon regulatory factor 3 (IRF3) in ALF mice, whereas the UT antagonist urantide inhibited the up-regulated liver IRF3. LPS stimulation induced IRF3 transcription and nuclear translocation and promoted the secretion of interleukin-6 (IL-6), interferon (IFN)-β, and IFN-γ in Kupffer cells (KCs); these effects in LPS-stimulated KCs were inhibited by urantide. Knockdown of IRF3 using an adenovirus expressing an IRF3 shRNA inhibited IFN-β transcription and secretion as well as tumor necrosis factor (TNF)-α and IL-1β secretion from LPS-stimulated KCs; additionally, IL-10 transcription and secretion were promoted in response to LPS. However, LPS-stimulated TNF-α and IL-1β mRNA was not affected in the KCs. The IRF3 shRNA also did not have a significant effect on the NF-κB p65 subunit and p38MAPK protein phosphorylation levels in the nuclei of LPS-stimulated KCs. Therefore, IRF3 expression and activation depended on the signal transduction of the UII/UT system, and played important roles in UII/UT-mediated immune inflammatory injury in the liver but did not affect NF-κB and p38 MAPK activity.
Collapse
Affiliation(s)
- Liang-ming Liu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Wen-juan Tu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Tong Zhu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Xiao-ting Wang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Zhi-li Tan
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Huan Zhong
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - De-yong Gao
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| | - Dong-yu Liang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People's Hospital Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
22
|
Cadirci E, Halici Z, Yayla M, Toktay E, Bayir Y, Karakus E, Topcu A, Buyuk B, Albayrak A. Blocking of urotensin receptors as new target for treatment of carrageenan induced inflammation in rats. Peptides 2016; 82:35-43. [PMID: 27208703 DOI: 10.1016/j.peptides.2016.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
This study investigated possible role of U-II and its receptor expression in inflammation by using UTR agonist and antagonist in carrageenan induced acute inflammation. Rats were divided into 5 groups as (1) Healthy control, (2) Carrageenan control, (3) Carrageenan +Indomethacin 20mg/kg, orally, (4) Carrageenan +AC7954 (U-II receptor agonist, intraperitoneally) 30mg/kg and (5) Carrageenan +SB657510 (UTR antagonist, intraperitoneally) 30mg/kg. 1h after drug administration, carrageenan was injected. At the 3rd hour after carrageenan injection, agonist produced no effect while antagonist 63% anti-inflammatory effect respectively. UTR and UT-II expression increased in carrageenan induced paw tissue. Antagonist administration prevented the decrease in an antioxidant system and also capable to decrease TNF-α and IL-6 mRNA expressions. This study showed the role of urotensin II receptors in the physiopathogenesis of acute inflammatory response that underlying many diseases accompanied by inflammation.
Collapse
Affiliation(s)
- Elif Cadirci
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Zekai Halici
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey.
| | - Muhammed Yayla
- Department of Pharmacology, Kafkas University Faculty of Medicine, 36240 Kars, Turkey
| | - Erdem Toktay
- Department of Histology and Embryology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| | - Yasin Bayir
- Department of Biochemistry, Ataturk University Faculty of Pharmacy, 25240 Erzurum, Turkey
| | - Emre Karakus
- Department of Pharmacology and Toxicology, Ataturk University Faculty of Veterinary Medicine, 25240 Erzurum, Turkey
| | - Atilla Topcu
- Department of Pharmacology, RTE University Faculty of Medicine, 53240 Rize, Turkey
| | - Basak Buyuk
- Department of Histology and Embryology, 19 Mart University, Faculty of Medicine, 25240 Çanakkale, Turkey
| | - Abdulmecit Albayrak
- Department of Pharmacology, Ataturk University Faculty of Medicine, 25240 Erzurum, Turkey
| |
Collapse
|
23
|
Luo SY, Chen S, Qin YD, Chen ZW. Urotensin-ⅡReceptor Antagonist SB-710411 Protects Rat Heart against Ischemia-Reperfusion Injury via RhoA/ROCK Pathway. PLoS One 2016; 11:e0146094. [PMID: 26771557 PMCID: PMC4714846 DOI: 10.1371/journal.pone.0146094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
Abstract
Aim SB-710411 is a rat selective urotensin-II (U-II) receptor antagonist, which can block U-II-induced contraction of the aorta and inhibit U-II-induced myocardial fibrosis in rats. However, the effect of SB-710411 on myocardial ischemia-reperfusion (I/R) injury is unclear. The present study was designed to investigate whether SB-710411 has a protective effect on myocardial I/R injury in rats and the possible mechanisms. Methods and Results Myocardial I/R injury was induced by occluding the left anterior descending coronary artery in adult male Sprague-Dawley rats. Hemodynamic parameters, electrocardiogram (ECG), infarct size, histological alteration, lactate dehydrogenase (LDH), creatine phosphokinase-MB (CK-MB), cardiac troponin I (cTnI), RhoA, and the protein expressions of U-II receptor (UTR), ROCK1 and ROCK2 were evaluated. Cardiac I/R injury significantly up-regulated the expressions of UTR, ROCK1 and ROCK2 proteins in rat myocardium. SB-710411 1.0 and 2.0 μg/kg significantly reduced cardiac I/R-induced the infarct size and histological damage in rat myocardium, markedly inhibited the changes of hemodynamic parameters and the increases of ST-segment in ECG, the serum LDH and CK-MB activities and cTnI level in rats subjected to myocardial I/R injury. Furthermore, SB-710411 obviously prevented myocardial I/R-increased RhoA activity and UTR, ROCK1 and ROCK2 protein expressions. Conclusions Our results indicate that cardiac I/R injury increases myocardial UTR expression, and SB-710411 has a potent protective effect on myocardial I/R injury in rats. The cardioprotection may be associated with the inhibition of UTR-RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Sheng-Yong Luo
- Department of Pharmacology, Anhui Medical University, Hefei, Anhui, China.,Anhui academy of medical sciences, Hefei, Anhui, China
| | - Shuo Chen
- Xinglin College of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yi-De Qin
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui, China
| | - Zhi-Wu Chen
- Department of Pharmacology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
24
|
Zhao S, Li Y, Gao S, Wang X, Sun L, Cheng D, Bai L, Guan H, Wang R, Fan J, Liu E. Autocrine Human Urotensin II Enhances Macrophage-Derived Foam Cell Formation in Transgenic Rabbits. BIOMED RESEARCH INTERNATIONAL 2015; 2015:843959. [PMID: 26640798 PMCID: PMC4659961 DOI: 10.1155/2015/843959] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/29/2015] [Indexed: 11/18/2022]
Abstract
Circulating urotensin II (UII) is involved in the development of atherosclerosis. However, the role of autocrine UII in the development of atherosclerosis remains unclear. Here, we tested the hypothesis that autocrine UII would promote atherosclerosis. Transgenic rabbits were created as a model to study macrophage-specific expressing human UII (hUII) and used to investigate the role of autocrine UII in the development of atherosclerosis. Transgenic rabbits and their nontransgenic littermates were fed a high cholesterol diet to induce atherosclerosis. Comparing the transgenic rabbits with their nontransgenic littermates, it was observed that hUII expression increased the macrophage-positive area in the atherosclerotic lesions by 45% and the positive area ratio by 56% in the transgenic rabbits. Autocrine hUII significantly decreased the smooth muscle cell-positive area ratio in transgenic rabbits (by 54%), without affecting the plasma levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, and glucose and adipose tissue contents. These results elucidated for the first time that autocrine UII plays an important role in the development of atherosclerosis by increasing the accumulation of macrophage-derived foam cell.
Collapse
Affiliation(s)
- Sihai Zhao
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Yafeng Li
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Shoucui Gao
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Xiaojing Wang
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Lijing Sun
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Daxing Cheng
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Liang Bai
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Hua Guan
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Rong Wang
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Enqi Liu
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi 710061, China
- Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| |
Collapse
|
25
|
Lim CJ, Jang JY, Kim SH, Lee BH, Oh KS, Yi KY. 1,3,4-Thiadiazol-2-amine Derivatives as Urotensin-II Receptor (UT) Antagonists. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Chae Jo Lim
- Bio & Drug Discovery Division; Korea Research Institute of Chemical Technology; Daejeon 34114 Korea
- Department of Medicinal Chemistry and Pharmacology; University of Science and Technology; Daejeon 34113 Korea
| | - Ju Young Jang
- Bio & Drug Discovery Division; Korea Research Institute of Chemical Technology; Daejeon 34114 Korea
- Department of Medicinal Chemistry and Pharmacology; University of Science and Technology; Daejeon 34113 Korea
| | - Sung Hwan Kim
- Bio & Drug Discovery Division; Korea Research Institute of Chemical Technology; Daejeon 34114 Korea
- Department of Medicinal Chemistry and Pharmacology; University of Science and Technology; Daejeon 34113 Korea
| | - Byung Ho Lee
- Bio & Drug Discovery Division; Korea Research Institute of Chemical Technology; Daejeon 34114 Korea
| | - Kwang-Seok Oh
- Bio & Drug Discovery Division; Korea Research Institute of Chemical Technology; Daejeon 34114 Korea
- Department of Medicinal Chemistry and Pharmacology; University of Science and Technology; Daejeon 34113 Korea
| | - Kyu Yang Yi
- Bio & Drug Discovery Division; Korea Research Institute of Chemical Technology; Daejeon 34114 Korea
- Department of Medicinal Chemistry and Pharmacology; University of Science and Technology; Daejeon 34113 Korea
| |
Collapse
|
26
|
Urotensin II Protects Cardiomyocytes from Apoptosis Induced by Oxidative Stress through the CSE/H2S Pathway. Int J Mol Sci 2015; 16:12482-98. [PMID: 26047336 PMCID: PMC4490456 DOI: 10.3390/ijms160612482] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 02/07/2023] Open
Abstract
Plasma urotensin II (UII) has been observed to be raised in patients with acute myocardial infarction; suggesting a possible cardiac protective role for this peptide. However, the molecular mechanism is unclear. Here, we treated cultured cardiomyocytes with H2O2 to induce oxidative stress; observed the effect of UII on H2O2-induced apoptosis and explored potential mechanisms. UII pretreatment significantly reduced the number of apoptotic cardiomyocytes induced by H2O2; and it partly abolished the increase of pro-apoptotic protein Bax and the decrease of anti-apoptotic protein Bcl-2 in cardiomyocytes induced by H2O2. SiRNA targeted to the urotensin II receptor (UT) greatly inhibited these effects. Further analysis revealed that UII increased the production of hydrogen sulfide (H2S) and the level of cystathionine-γ-lyase (CSE) by activating the ERK signaling in H2O2-treated-cardiomyocytes. Si-CSE or ERK inhibitor not only greatly inhibited the increase in CSE level or the phosphorylation of ERK induced by UII but also reversed anti-apoptosis of UII in H2O2-treated-cadiomyocytes. In conclusion, UII rapidly promoted the phosphorylation of ERK and upregulated CSE level and H2S production, which in turn activated ERK signaling to protect cardiomyocytes from apoptosis under oxidative stress. These results suggest that increased plasma UII level may protect cardiomyocytes at the early-phase of acute myocardial infarction in patients.
Collapse
|
27
|
Oh KS, Lee JH, Yi KY, Lim CJ, Lee S, Park CH, Seo HW, Lee BH. The orally active urotensin receptor antagonist, KR36676, attenuates cellular and cardiac hypertrophy. Br J Pharmacol 2015; 172:2618-33. [PMID: 25597918 DOI: 10.1111/bph.13082] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/11/2014] [Accepted: 01/13/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Blockade of the actions of urotensin-II (U-II) mediated by the urotensin (UT) receptor should improve cardiac function and prevent cardiac remodelling in cardiovascular disease. Here, we have evaluated the pharmacological properties of the recently identified UT receptor antagonist, 2-(6,7-dichloro-3-oxo-2H-benzo[b][1,4]oxazin-4(3H)-yl)-N-methyl-N-(2-(pyrrolidin-1-yl)-1-(4-(thiophen-3-yl)phenyl) ethyl)acetamide (KR36676). EXPERIMENTAL APPROACH Pharmacological properties of KR36676 were studied in a range of in vitro assays (receptor binding, calcium mobilization, stress fibre formation, cellular hypertrophy) and in vivo animal models such as cardiac hypertrophy induced by transverse aortic constriction (TAC) or myocardial infarction (MI). KEY RESULTS KR36676 displayed high binding affinity for the UT receptor (Ki : 0.7 nM), similar to that of U-II (0.4 nM), and was a potent antagonist at that receptor (IC50 : 4.0 nM). U-II-induced stress fibre formation and cellular hypertrophy were significantly inhibited with low concentrations of KR36676 (≥0.01 μM). Oral administration of KR36676 (30 mg·kg(-1) ) in a TAC model in mice attenuated cardiac hypertrophy and myocardial fibrosis. Moreover, KR36676 restored cardiac function and myocyte size in rats with MI-induced cardiac hypertrophy. CONCLUSIONS AND IMPLICATIONS A highly potent UT receptor antagonist exerted anti-hypertrophic effects not only in infarcted rat hearts but also in pressure-overloaded mouse hearts. KR36676 could be a valuable pharmacological tool in elucidating the complicated physiological role of U-II and UT receptors in cardiac hypertrophy.
Collapse
Affiliation(s)
- K S Oh
- Research Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon, Korea; Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, Daejeon, Korea
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Liu LM, Liang DY, Ye CG, Tu WJ, Zhu T. The UII/UT system mediates upregulation of proinflammatory cytokines through p38 MAPK and NF-κB pathways in LPS-stimulated Kupffer cells. PLoS One 2015; 10:e0121383. [PMID: 25803040 PMCID: PMC4372515 DOI: 10.1371/journal.pone.0121383] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/31/2015] [Indexed: 01/27/2023] Open
Abstract
The urotensin II (UII)/UII receptor (UT) system is closely related to immune inflammation. In acute liver failure (ALF), the UII/UT system can promote the production and release of proinflammatory cytokines, inducing an inflammatory injury response in liver tissue. However, the mechanism by which the hepatic UII/UT system promotes proinflammatory cytokine production and release is not clear. To solve this problem, we used primary Kupffer cells (KCs) as the model system in the current study. The results showed that after lipopolysaccharide (LPS) stimulation, KCs showed significantly increased expression and release of UII/UT and proinflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 1β (IL-1β). Pretreatment with urantide, which is a UT receptor antagonist, significantly inhibited the LPS-stimulated expression and release of UII/UT, TNF-α, and IL-1β by KCs. In addition, LPS stimulation induced nuclear p38 mitogen-activated protein kinase (MAPK) protein phosphorylation and expression of the nuclear nuclear factor κB (NF-κB) p65 subunit in KCs and enhanced the binding activity of NF-κB to DNA molecules, whereas urantide pretreatment significantly inhibited the LPS-stimulated nuclear expression and activity of these molecules in KCs. Therefore, our conclusion is that the UII/UT system mediates LPS-stimulated production and release of proinflammatory cytokine by KCs, and this mediating effect at least partially relies on the inflammatory signaling pathway molecules p38 MAPK and NF-κB.
Collapse
Affiliation(s)
- Liang Ming Liu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Dong Yu Liang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Chang Gen Ye
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Wen Juan Tu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Tong Zhu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
29
|
Vaudry H, Leprince J, Chatenet D, Fournier A, Lambert DG, Le Mével JC, Ohlstein EH, Schwertani A, Tostivint H, Vaudry D. International Union of Basic and Clinical Pharmacology. XCII. Urotensin II, urotensin II-related peptide, and their receptor: from structure to function. Pharmacol Rev 2015; 67:214-58. [PMID: 25535277 DOI: 10.1124/pr.114.009480] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Urotensin II (UII) is a cyclic neuropeptide that was first isolated from the urophysis of teleost fish on the basis of its ability to contract the hindgut. Subsequently, UII was characterized in tetrapods including humans. Phylogenetic studies and synteny analysis indicate that UII and its paralogous peptide urotensin II-related peptide (URP) belong to the somatostatin/cortistatin superfamily. In mammals, the UII and URP genes are primarily expressed in cholinergic neurons of the brainstem and spinal cord. UII and URP mRNAs are also present in various organs notably in the cardiovascular, renal, and endocrine systems. UII and URP activate a common G protein-coupled receptor, called UT, that exhibits relatively high sequence identity with somatostatin, opioid, and galanin receptors. The UT gene is widely expressed in the central nervous system (CNS) and in peripheral tissues including the retina, heart, vascular bed, lung, kidney, adrenal medulla, and skeletal muscle. Structure-activity relationship studies and NMR conformational analysis have led to the rational design of a number of peptidic and nonpeptidic UT agonists and antagonists. Consistent with the wide distribution of UT, UII has now been shown to exert a large array of biologic activities, in particular in the CNS, the cardiovascular system, and the kidney. Here, we review the current knowledge concerning the pleiotropic actions of UII and discusses the possible use of antagonists for future therapeutic applications.
Collapse
Affiliation(s)
- Hubert Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jérôme Leprince
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Chatenet
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Alain Fournier
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David G Lambert
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jean-Claude Le Mével
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Eliot H Ohlstein
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Adel Schwertani
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Hervé Tostivint
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| |
Collapse
|
30
|
Lim CJ, Oh SA, Lee BH, Oh KS, Yi KY. Synthesis and SAR of thieno[3,2- b ]pyridinyl urea derivatives as urotensin-II receptor antagonists. Bioorg Med Chem Lett 2014; 24:5832-5835. [DOI: 10.1016/j.bmcl.2014.09.089] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 02/07/2023]
|
31
|
Hwang HJ, Jung TW, Ryu JY, Hong HC, Choi HY, Seo JA, Kim SG, Kim NH, Choi KM, Choi DS, Baik SH, Yoo HJ. Dipeptidyl petidase-IV inhibitor (gemigliptin) inhibits tunicamycin-induced endoplasmic reticulum stress, apoptosis and inflammation in H9c2 cardiomyocytes. Mol Cell Endocrinol 2014; 392:1-7. [PMID: 24813659 DOI: 10.1016/j.mce.2014.04.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/07/2014] [Accepted: 04/29/2014] [Indexed: 01/05/2023]
Abstract
The direct effects of dipeptidyl peptidase-IV (DPP-IV) inhibitors on endoplasmic reticulum (ER) stress-induced apoptosis and inflammation in cardiomyocytes have not been elucidated. H9c2 cell viability, which was reduced by tunicamycin, was increased after DPP-IV inhibitor gemigliptin treatment. Gemigliptin significantly decreased the tunicamycin-mediated increase in glucose regulated protein 78 (GRP78) expression and ER stress-mediated signaling molecules such as protein kinase RNA-like endoplasmic reticulum kinase (PERK)/C-EBP homologous protein (CHOP) and inositol-requiring enzyme 1α (IRE1α)/c-Jun N-terminal kinase (JNK)-p38. Furthermore, gemigliptin effectively induced Akt phosphorylation in a dose-dependent manner. Using flow cytometry and Hoechst staining, we showed that treatment with Akt inhibitor significantly blocked the anti-apoptotic effects mediated by gemigliptin. The reduction in tunicamycin-induced GRP78 level and PERK/CHOP pathway activity by gemigliptin was reversed after treatment with Akt inhibitor. In conclusion, gemigliptin effectively inhibited ER stress-induced apoptosis and inflammation in cardiomyocytes via Akt/PERK/CHOP and IRE1α/JNK-p38 pathways, suggesting its direct protective role in cardiovascular diseases.
Collapse
Affiliation(s)
- Hwan-Jin Hwang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ja Young Ryu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ho Cheol Hong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hae Yoon Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Dong Seop Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hye Jin Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Gögebakan B, Uruc V, Ozden R, Duman IG, Yagiz AE, Okuyan HM, Aldemir O, Dogramaci Y, Kalaci A. Urotensin II (U-II), a novel cyclic peptide, possibly associated with the pathophysiology of osteoarthritis. Peptides 2014; 54:159-61. [PMID: 24468547 DOI: 10.1016/j.peptides.2014.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/16/2014] [Accepted: 01/16/2014] [Indexed: 01/27/2023]
Abstract
Synovial fibrosis is one of the main outcomes of osteoarthritis. Some authors have reported that urotensin-II (U-II) may cause pathologic fibrosis in cardiovascular system, lung and liver. However there are no previous reports available in the literature about its relationship with the synovial fibrosis in osteoarthritis. The aim of this study was to compare the U-II levels in knee synovial fluids obtained from osteoarthritic and non-osteoarthritic patients. Two groups were created, the osteoarthritis group and non-osteoarthritic control group. The control group was consisted of patients who underwent arthroscopic surgery for other reasons than cartilage disorders. In the osteoarthritis group all patients had grade 4 primer degenerative osteoarthritis and were treated with total knee arthroplasty. Minimum 1 mL knee synovial fluids were obtained during operation. Levels of U-II were measured by using ELISA kit U-II levels were significantly higher in the osteoarthritic group than that in the control group. No correlation was found between U-II levels and age. In conclusion, the significantly high U-II levels in the knee synovial fluid of osteoarthritic patients supported our hypothesis that "U-II may be associated with the synovial fibrosis in osteoarthritis".
Collapse
Affiliation(s)
- Bülent Gögebakan
- Mustafa Kemal University, Faculty of Medicine, Department of Medical Biology, 31034 Hatay, Turkey.
| | - Vedat Uruc
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Raif Ozden
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Ibrahim Gokhan Duman
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Abdullah Erman Yagiz
- Mustafa Kemal University, Faculty of Medicine, Department of Physical Therapy and Rehabilitation, 31034 Hatay, Turkey
| | - Hamza Malik Okuyan
- Mustafa Kemal University, Faculty of Medicine, Department of Medical Biology, 31034 Hatay, Turkey
| | - Ozgur Aldemir
- Mustafa Kemal University, Faculty of Medicine, Department of Medical Biology, 31034 Hatay, Turkey
| | - Yunus Dogramaci
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| | - Aydiner Kalaci
- Mustafa Kemal University, Faculty of Medicine, Department of Orthopedics and Traumatology, 31034 Hatay, Turkey
| |
Collapse
|
33
|
Zhao J, Zhang SF, Shi Y, Ren LQ. Effects of urotensin II and its specific receptor antagonist urantide on rat vascular smooth muscle cells. Bosn J Basic Med Sci 2014; 13:78-83. [PMID: 23725502 DOI: 10.17305/bjbms.2013.2369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We investigated the effects of urantide, a receptor antagonist of urotensin II (U-II), on the expression of U-II and its receptor GPR14 in rat vascular smooth muscle cells. Vascular smooth muscle cells from rat thoracic aorta were cultured by explant method. Subjects in this experiment were divided into eight groups: normal control group (group C), U-II group (group M), positive control group (Flu group) and urantide-treated groups (10⁻¹⁰, 10⁻⁹, 10⁻⁸, 10⁻⁷ and 10⁻⁶ mol/L). Cultured vascular smooth muscle cells in vitro were studied by immunocytochemistry, biochemistry, and flow cytometry. U-II (10⁻⁸ mol/L) promoted the proliferation of vascular smooth muscle cells at each time point, influenced cell cycle, increased proliferation index and S-phase cell fraction, and dramatically promoted the expression of U-II and GPR14. In the concentration range from 10⁻¹⁰ to 10⁻⁶ mol/L, urantide dramatically inhibited the proliferation of vascular smooth muscle cells and the protein expression of U-II and GPR14, especially at a concentration of 10⁻⁶ mol/L. U-II, binding with its receptor GPR14, promotes vascular smooth muscle cells proliferation and migration, which can be inhibited by urantide. This study provides an evidence for understanding the effects of U-II and its receptor GPR14 on vascular smooth muscle cells.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Pathophysiology, Chengde Medical College, Chengde 067000, China
| | | | | | | |
Collapse
|
34
|
Chen Z, Xu J, Ye Y, Li Y, Gong H, Zhang G, Wu J, Jia J, Liu M, Chen Y, Yang C, Tang Y, Zhu Y, Ge J, Zou Y. Urotensin II inhibited the proliferation of cardiac side population cells in mice during pressure overload by JNK-LRP6 signalling. J Cell Mol Med 2014; 18:852-62. [PMID: 24447593 PMCID: PMC4119391 DOI: 10.1111/jcmm.12230] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 12/09/2013] [Indexed: 01/31/2023] Open
Abstract
Cardiac side population cells (CSPs) are promising cell resource for the regeneration in diseased heart as intrinsic cardiac stem cells. However, the relative low ratio of CSPs in the heart limited the ability of CSPs to repair heart and improve cardiac function effectively under pathophysiological condition. Which factors limiting the proliferation of CSPs in diseased heart are unclear. Here, we show that urotensin II (UII) regulates the proliferation of CSPs by c-Jun N-terminal kinase (JNK) and low density lipoprotein receptor-related protein 6 (LRP6) signalling during pressure overload. Pressure overload greatly upregulated UII level in plasma, UII receptor (UT) antagonist, urantide, promoted CSPs proliferation and improved cardiac dysfunction during chronic pressure overload. In cultured CSPs subjected to mechanical stretch (MS), UII significantly inhibited the proliferation by UT. Nanofluidic proteomic immunoassay showed that it is the JNK activation, but not the extracellular signal-regulated kinase signalling, that involved in the UII-inhibited- proliferation of CSPs during pressure overload. Further analysis in vitro indicated UII-induced-phospho-JNK regulates phosphorylation of LRP6 in cultured CSPs after MS, which is important in the inhibitory effect of UII on the CSPs during pressure overload. In conclusion, UII inhibited the proliferation of CSPs by JNK/LRP6 signalling during pressure overload. Pharmacological inhibition of UII promotes CSPs proliferation in mice, offering a possible therapeutic approach for cardiac failure induced by pressure overload.
Collapse
Affiliation(s)
- Zhidan Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Plasma Urotensin II Act as a Diagnostic Biomarker for Acute Coronary Syndromes. Int J Pept Res Ther 2013. [DOI: 10.1007/s10989-013-9376-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Early growth response 1 is an early signal inducing Cav3.2 T-type calcium channels during cardiac hypertrophy. Cardiovasc Res 2013; 100:222-30. [DOI: 10.1093/cvr/cvt190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
37
|
Liang DY, Liu LM, Ye CG, Zhao L, Yu FP, Gao DY, Wang YY, Yang ZW, Wang YY. Inhibition of UII/UTR system relieves acute inflammation of liver through preventing activation of NF-κB pathway in ALF mice. PLoS One 2013; 8:e64895. [PMID: 23755157 PMCID: PMC3670940 DOI: 10.1371/journal.pone.0064895] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/19/2013] [Indexed: 02/06/2023] Open
Abstract
Urotensin II (UII) is implicated in immune inflammatory diseases through its specific high-affinity UT receptor (UTR). Enhanced expression of UII/UTR was recently demonstrated in the liver with acute liver failure (ALF). Here, we analysed the relationship between UII/UTR expression and ALF in lipopolysaccharide (LPS)/D-galactosamine (GalN)-challenged mice. Thereafter, we investigated the effects produced by the inhibition of UII/UTR system using urantide, a special antagonist of UTR, and the potential molecular mechanisms involved in ALF. Urantide was administered to mice treated with LPS/GalN. Expression of UII/UTR, releases of proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β) and interferon-γ (IFN-γ), and activation of nuclear factor κB (NF-κB) signaling pathway were assessed in the lethal ALF with or without urantide pretreatment. We found that LPS/GalN-challenged mice showed high mortality and marked hepatic inflammatory infiltration and cell apoptosis as well as a significant increase of UII/UTR expression. Urantide pretreatment protected against the injury in liver following downregulation of UII/UTR expression. A close relationship between the acutely flamed hepatic injury and UII/UTR expression was observed. In addition, urantide prevented the increases of proinflammatory cytokines such as TNF-α, IL-1β and IFN-γ, and activation of NF-κB signaling pathway induced by LPS/GalN in mice. Thus, we conclude that UII/UTR system plays a role in LPS/GalN-induced ALF. Urantide has a protective effect on the acutely inflamed injury of liver in part through preventing releases of proinflammatory cytokines and activation of NF-κB pathway.
Collapse
Affiliation(s)
- Dong-yu Liang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Liang-ming Liu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- Department of Infection, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Chang-gen Ye
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- Department of Infection, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Liang Zhao
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Fang-ping Yu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - De-yong Gao
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- Department of Infection, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Ying-ying Wang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Zhi-wen Yang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Yan-yan Wang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
38
|
Zhao J, Yu QX, Kong W, Gao HC, Sun B, Xie YQ, Ren LQ. The urotensin II receptor antagonist, urantide, protects against atherosclerosis in rats. Exp Ther Med 2013; 5:1765-1769. [PMID: 23837070 PMCID: PMC3702698 DOI: 10.3892/etm.2013.1052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/05/2013] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to explore the use of urantide as an antagonist of the urotensin II (UII) receptor, G protein-coupled receptor 14 (GPR14), to protect against atherosclerosis (AS) in rats. The AS rat model was induced by an intraperitoneal injection of vitamin D3 (VD3) into rats fed with a high-fat diet for four weeks. Urantide was then injected into the rats. Immunohistochemical staining, serum biochemical assay, reverse transcription-polymerase chain reaction (RT-PCR) and western blotting were used to investigate the expression of UII and its receptor GPR14 in the AS rat model. Four weeks after induction, pathological changes typical of AS were observed in the AS rat model. In the plaques of the aortic tunica intima and tunica media, expression of UII and GPR14 was observed. The protein and gene expression levels of UII and GPR14 in the model group were significantly increased compared with those in the normal group (P<0.01). Urantide ameliorated the pathological changes of AS in the rat model and reduced the gene and protein expression levels of UII and GPR14 (P<0.05 or P<0.01). UII is associated with AS and the UII receptor GPR14-specific antagonist, urantide, demonstrates the ability to protect against AS. Thus, this study provides new insight and experimental theories for the clinical application of urantide to treat AS.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000
| | | | | | | | | | | | | |
Collapse
|
39
|
Ames RS, Lu Q. Viral-mediated gene delivery for cell-based assays in drug discovery. Expert Opin Drug Discov 2013; 4:243-56. [PMID: 23489124 DOI: 10.1517/17460440902751599] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Adenovirus, retrovirus and lentivirus-based vectors, originally engineered and optimized for in vivo and ex vivo gene therapy, have become increasingly useful for viral-mediated gene delivery to support in vitro cell-based assays. Viral vectors underpin functional genomics screening of cDNA, shRNA and aptamer libraries, are used for a variety of target validation studies and importantly, for high-throughput cell-based drug discovery and compound profiling assays. The baculovirus/insect cell expression system had gained prevalence as a tool for recombinant protein production when it was observed that recombinant baculovirus vectors too could serve as efficient gene delivery vehicles for a wide range of mammalian cells. Although the use of baculovirus vectors in vivo has lagged behind retroviral, adenoviral and lentiviral vectors, they have gained prominence for development of in vitro cell-based assays due to the ease of generation, broad host range and excellent biosafety profile. There is an increasing emphasis on cell-based assays in high-throughput automated drug discovery laboratories and a variety of commercially available viral-vectors can be used for supporting these assays. OBJECTIVE We compare and contrast the current viral-mediated gene delivery vector systems and highlight their suitability for cell-based drug discovery assays. CONCLUSION Viral-mediated gene delivery is increasingly being used in support of genome scale target validation studies and cell-based assay development for specific drug target genes such as ion channels, G protein-coupled receptors and intracellular enzymes. The choice of a delivery system over another for a particular application is largely dictated by the cell types and cell lines in use, virus cellular tropism, assay throughput, safety requirements and ease/cost of reagent generation.
Collapse
Affiliation(s)
- Robert S Ames
- Molecular Discovery Research, GlaxoSmithKline R&D, UE0433, 709 Swedeland Road, King of Prussia, PA 19406, USA +1 610 270 7602 ; +1 610 270 7359 ;
| | | |
Collapse
|
40
|
Zhou CH, Wan YY, Chu XH, Song Z, Xing SH, Wu YQ, Yin XX. Urotensin II contributes to the formation of lung adenocarcinoma inflammatory microenvironment through the NF-κB pathway in tumor-bearing nude mice. Oncol Lett 2012; 4:1259-1263. [PMID: 23226801 DOI: 10.3892/ol.2012.932] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 09/04/2012] [Indexed: 12/29/2022] Open
Abstract
Urotensin II (UII), a somatostatin-like cyclic peptide, was originally isolated from the fish urophysis. Our previous study showed that UII stimulates the proliferation of A549 lung adenocarcinoma cells and promotes tumor growth in a nude mouse xenograft model, suggesting that UII may contribute to the pathogenesis of lung adenocarcinoma. In this study, the underlying mechanism for UII to promote lung adenocarcinoma growth was explored by observing the effect of UII on the tumor inflammatory microenvironment in tumor-bearing nude mice. Immunohistochemical analysis showed that UII promoted the infiltration of CD68(+) tumor-associated macrophages (TAMs) in the tumor micro-environment. Enzyme-linked immunosorbent assay (ELISA) demonstrated that UII promoted the release of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and matrix metalloproteinase-9 (MMP-9). Western blot analysis showed that UII promoted the activation of nuclear factor-κB (NF-κB). These findings suggest that the enhanced levels of IL-6, TNF-α and MMP-9 in the tumor microenvironment, which likely resulted from increased activation of NF-κB induced by UII, may be one of the important mechanisms by which UII promotes lung adenocarcinoma growth. These findings imply that antagonists of UII or urotensin II-receptor (UT-R) have potential for the prevention and treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Cheng-Hua Zhou
- School of Pharmacy, Xuzhou Medical College, Xuzhou, Jiangsu 221004
| | | | | | | | | | | | | |
Collapse
|
41
|
Liu LM, Liang DY, Zhang FF, Yu FP, Zhao L, Ye CG. Expression and role of Urotensin Ⅱ in lipopolysaccharide/D-galactosamine-induced acute liver failure in mice. Shijie Huaren Xiaohua Zazhi 2012; 20:1616-1622. [DOI: 10.11569/wcjd.v20.i18.1616] [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
AIM: To investigate the expression and role of Urotensin Ⅱ (UⅡ) in lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced acute liver failure (ALF) in mice.
METHODS: Male Balb/c mice were randomly and equally divided into four groups: normal control group (group A), pre-treatment control group (group B), ALF model group (group C), and pre-treatment model group (group D). ALF were induced in mice by intraperitoneal injection of LPS (50 μg/kg body weight)/D-GalN (800 mg/kg body weight). The pre-treatment mice were intravenously injected with Urantide (0.6 mg/kg body weight) 30 min before model induction. Serum and liver tissues were sampled 12 h after LPS/D-GalN injection. Mortality was calculated 24 h after attack. Serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were detected using the Reitman-Frankel method. Histopathological changes were observed by hematoxylin and eosin (HE) staining. Serum UⅡ levels were assessed by ELISA, and the expression of UⅡ and UT was detected by RT-PCR and immunohistochemistry.
RESULTS: A mortality of 66.7% was observed in group C, while all mice of groups A, B and D survived. Serum ALT and AST levels had a dramatic increase in groups C and D, but were significantly lower in group D than in group C (2 271.09 U/L ± 102.24 U/L vs 1 160.67 U/L ± 258.32 U/L, 1 569.42 U/L ± 204.04 U/L vs 1 030.31 U/L ± 108.09 U/L, both P < 0.01). Widespread destruction of liver architecture, hemorrhagic necrosis, and neutrophil infiltration were noted in group C, whereas liver architecture was completely preserved, and focal necrosis and fewer neutrophil infiltrates were observed in group D. After LPS/D-GalN challenge, serum UⅡ levels increased sharply in groups C and D, but were lower in group D than in group C (3.73 μg/L ± 0.52 μg/L vs 1.90 μg/L ± 0.27 μg/L, both P < 0.01). Overexpression of liver UⅡ and UT mRNAs and proteins was induced by the injection of LPS/D-GalN in groups C and D. Compared to group C, group D had lower levels of UⅡ and UT in the liver.
CONCLUSION: UⅡ expression and secretion can be induced by LPS/D-GalN challenge in the liver tissue of ALF mice via a positive feedback mechanism associated with promoting the expression of its receptor UT. UⅡ/UT receptor may be a vital mediator of LPS/D-GalN-induced ALF.
Collapse
|
42
|
Desrues L, Lefebvre T, Lecointre C, Schouft MT, Leprince J, Compère V, Morin F, Proust F, Gandolfo P, Tonon MC, Castel H. Down-regulation of GABA(A) receptor via promiscuity with the vasoactive peptide urotensin II receptor. Potential involvement in astrocyte plasticity. PLoS One 2012; 7:e36319. [PMID: 22563490 PMCID: PMC3341351 DOI: 10.1371/journal.pone.0036319] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 04/02/2012] [Indexed: 02/07/2023] Open
Abstract
GABAA receptor (GABAAR) expression level is inversely correlated with the proliferation rate of astrocytes after stroke or during malignancy of astrocytoma, leading to the hypothesis that GABAAR expression/activation may work as a cell proliferation repressor. A number of vasoactive peptides exhibit the potential to modulate astrocyte proliferation, and the question whether these mechanisms may imply alteration in GABAAR-mediated functions and/or plasma membrane densities is open. The peptide urotensin II (UII) activates a G protein-coupled receptor named UT, and mediates potent vasoconstriction or vasodilation in mammalian vasculature. We have previously demonstrated that UII activates a PLC/PIPs/Ca2+ transduction pathway, via both Gq and Gi/o proteins and stimulates astrocyte proliferation in culture. It was also shown that UT/Gq/IP3 coupling is regulated by the GABAAR in rat cultured astrocytes. Here we report that UT and GABAAR are co-expressed in cerebellar glial cells from rat brain slices, in human native astrocytes and in glioma cell line, and that UII inhibited the GABAergic activity in rat cultured astrocytes. In CHO cell line co-expressing human UT and combinations of GABAAR subunits, UII markedly depressed the GABA current (β3γ2>α2β3γ2>α2β1γ2). This effect, characterized by a fast short-term inhibition followed by drastic and irreversible run-down, is not relayed by G proteins. The run-down partially involves Ca2+ and phosphorylation processes, requires dynamin, and results from GABAAR internalization. Thus, activation of the vasoactive G protein-coupled receptor UT triggers functional inhibition and endocytosis of GABAAR in CHO and human astrocytes, via its receptor C-terminus. This UII-induced disappearance of the repressor activity of GABAAR, may play a key role in the initiation of astrocyte proliferation.
Collapse
Affiliation(s)
- Laurence Desrues
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Thomas Lefebvre
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Céline Lecointre
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Marie-Thérèse Schouft
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Jérôme Leprince
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Vincent Compère
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
- Department of Anesthesiology and Critical Care, Rouen University Hospital, Rouen, France
| | - Fabrice Morin
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - François Proust
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
- Department of Neurosurgery, Rouen University Hospital, Rouen, France
| | - Pierrick Gandolfo
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Marie-Christine Tonon
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
| | - Hélène Castel
- Inserm U982, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Astrocyte and Vascular Niche, University of Rouen, Mont-Saint-Aignan, France
- Institute of Research and Biomedical Innovation (IRIB), Normandy University PRES, University of Rouen, Mont-Saint-Aignan, France
- * E-mail:
| |
Collapse
|
43
|
Chen YL, Loh SH, Chen JJ, Tsai CS. Urotensin II prevents cardiomyocyte apoptosis induced by doxorubicin via Akt and ERK. Eur J Pharmacol 2012; 680:88-94. [DOI: 10.1016/j.ejphar.2012.01.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 01/27/2012] [Accepted: 01/28/2012] [Indexed: 12/23/2022]
|
44
|
Barrette PO, Schwertani AG. A closer look at the role of urotensin II in the metabolic syndrome. Front Endocrinol (Lausanne) 2012; 3:165. [PMID: 23293629 PMCID: PMC3531708 DOI: 10.3389/fendo.2012.00165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/29/2012] [Indexed: 12/12/2022] Open
Abstract
Urotensin II (UII) is a vasoactive peptide that was first discovered in the teleost fish, and later in mammals and humans. UII binds to the G protein coupled receptor GPR14 (now known as UT). UII mediates important physiological and pathological actions by interacting with its receptor. The metabolic syndrome (MetS) is described as cluster of factors such as obesity, dyslipidemia, hypertension, and insulin resistance (IR), further leading to development of type 2 diabetes mellitus and cardiovascular diseases. UII levels are upregulated in patients with the MetS. Evidence directly implicating UII in every risk factor of the MetS has been accumulated. The mechanism that links the different aspects of the MetS relies primarily on IR and inflammation. By directly modulating both of these factors, UII is thought to play a central role in the pathogenesis of the MetS. Moreover, UII also plays an important role in hypertension and hyperlipidemia thereby contributing to cardiovascular complications associated with the MetS.
Collapse
Affiliation(s)
| | - Adel Giaid Schwertani
- *Correspondence: Adel Giaid Schwertani, Division of Cardiology, Department of Medicine, McGill University Health Center, 1650 Cedar Avenue, Room C9-166, Montreal, QC, Canada H3G 1A4. e-mail:
| |
Collapse
|
45
|
Tsoukas P, Kane E, Giaid A. Potential Clinical Implications of the Urotensin II Receptor Antagonists. Front Pharmacol 2011; 2:38. [PMID: 21811463 PMCID: PMC3143724 DOI: 10.3389/fphar.2011.00038] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Accepted: 07/05/2011] [Indexed: 12/20/2022] Open
Abstract
Urotensin II (UII) binds to its receptor, UT, playing an important role in the heart, kidneys, pancreas, adrenal gland, and central nervous system. In the vasculature, it acts as a potent endothelium-independent vasoconstrictor and endothelium-dependent vasodilator. In disease states, however, this constriction–dilation equilibrium is disrupted. There is an upregulation of the UII system in heart disease, metabolic syndrome, and kidney failure. The increase in UII release and UT expression suggest that UII system may be implicated in the pathology and pathogenesis of these diseases by causing an increase in acyl-coenzyme A:cholesterol acyltransferase-1 (ACAT-1) activity leading to smooth muscle cell proliferation and foam cell infiltration, insulin resistance (DMII), as well as inflammation, high blood pressure, and plaque formation. Recently, UT antagonists such as SB-611812, palosuran, and most recently a piperazino-isoindolinone based antagonist have been developed in the hope of better understanding the UII system and treating its associated diseases.
Collapse
Affiliation(s)
- Philip Tsoukas
- Division of Cardiology, Department of Medicine, Montreal General Hospital, McGill University Health Center Montreal, QC, Canada
| | | | | |
Collapse
|
46
|
Urotensin-2 promotes collagen synthesis via ERK1/2-dependent and ERK1/2-independent TGF-β1 in neonatal cardiac fibroblasts. Cell Biol Int 2011; 35:93-8. [PMID: 20946103 DOI: 10.1042/cbi20090104] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
U2 (urotensin-2) is the most potent vasoconstrictor in mammals which is involved in cardiac remodelling, including cardiac hypertrophy and cardiac fibrosis. Although the cellular mechanisms of the U2-induced vasoconstriction have been extensively studied, the signalling pathways involved in U2-induced TGF-β1 (transforming growth factor-β1) expression and collagen synthesis remain unclear. In this study, we show that U2 promoted collagen synthesis and ERK1/2 (extracellular signal-regulated kinase 1/2) activation in neonatal cardiac fibroblasts. The U2-induced collagen synthesis and TGF-β1 production were significantly but not completely inhibited by blocking ERK1/2. Both ERK1/2 inhibitor and TGF-β1 antibody could separately inhibit U2-induced collagen synthesis, and the synergistic inhibition effect was observed by blocking ERK1/2 and TGF-β1 simultaneously. These data suggest that U2 promotes collagen synthesis via ERK1/2-dependent and independent TGF-β1 pathway in neonatal cardiac fibroblasts.
Collapse
|
47
|
Gong H, Ma H, Liu M, Zhou B, Zhang G, Chen Z, Jiang G, Yan Y, Yang C, Kanda M, Wu J, Yuan J, Li L, Nagai T, Komuro I, Ge J, Zou Y. Urotensin II inhibits the proliferation but not the differentiation of cardiac side population cells. Peptides 2011; 32:1035-41. [PMID: 21291940 DOI: 10.1016/j.peptides.2011.01.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 01/22/2011] [Accepted: 01/24/2011] [Indexed: 11/26/2022]
Abstract
Urotensin II (UII) induces the development of cardiac remodeling and atherosclerosis by promoting hypertrophy of cardiomyocytes and mitogenesis of fibroblasts and vascular smooth muscle cells. But its effect on cardiac side population cells (CSPs), one of somatic stem cells, is unclear. The present study examined the influences of UII on the differentiation and proliferation of CSPs. CSPs were isolated from neonatal rat hearts by fluorescence-activated cell sorting (FACS) and cultured with or without the presence of UII (10(-8), 10(-7), 10(-6)mol/l). The expressions of α-cardiac myosin heavy chain (α-MHC), α-smooth muscle actin (SMA) and Von Willebrand factor (vWF) mRNAs and proteins were analyzed by reverse transcriptional PCR (RT-PCR) and immunofluorescence to evaluate the differentiation of CSPs into cardiomyocytes, smooth muscle cells and endothelial cells, respectively. The proliferation of CSPs was assessed by Luminescent Cell Viability Assay. The influence of UII on the proliferation of CSPs in vivo was also evaluated by FACS. Our results revealed that UII did inhibit the proliferation of CSPs through up-regulation of phosphorylated c-Jun N-terminal protein kinase (JNK), although it didn't affect the differentiation of cultured CSPs. Experiments in vivo also showed that UII reduced the number of CSPs in mice compared with control group. These data indicate that UII reduces the number of CSPs by inhibiting the proliferation of CSPs possibly through increase of JNK phosphorylation.
Collapse
Affiliation(s)
- Hui Gong
- Institutes of Biomedical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Behm DJ, Aiyar NV, Olzinski AR, McAtee JJ, Hilfiker MA, Dodson JW, Dowdell SE, Wang GZ, Goodman KB, Sehon CA, Harpel MR, Willette RN, Neeb MJ, Leach CA, Douglas SA. GSK1562590, a slowly dissociating urotensin-II receptor antagonist, exhibits prolonged pharmacodynamic activity ex vivo. Br J Pharmacol 2010; 161:207-28. [PMID: 20718751 DOI: 10.1111/j.1476-5381.2010.00889.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Recently identified antagonists of the urotensin-II (U-II) receptor (UT) are of limited utility for investigating the (patho)physiological role of U-II due to poor potency and limited selectivity and/or intrinsic activity. EXPERIMENTAL APPROACH The pharmacological properties of two novel UT antagonists, GSK1440115 and GSK1562590, were compared using multiple bioassays. KEY RESULTS GSK1440115 (pK(i)= 7.34-8.64 across species) and GSK1562590 (pK(i)= 9.14-9.66 across species) are high affinity ligands of mammalian recombinant (mouse, rat, cat, monkey, human) and native (SJRH30 cells) UT. Both compounds exhibited >100-fold selectivity for UT versus 87 distinct mammalian GPCR, enzyme, ion channel and neurotransmitter uptake targets. GSK1440115 showed competitive antagonism at UT in arteries from all species tested (pA(2)= 5.59-7.71). In contrast, GSK1562590 was an insurmountable UT antagonist in rat, cat and hUT transgenic mouse arteries (pK(b)= 8.93-10.12 across species), but a competitive antagonist in monkey arteries (pK(b)= 8.87-8.93). Likewise, GSK1562590 inhibited the hU-II-induced systemic pressor response in anaesthetized cats at a dose 10-fold lower than that of GSK1440115. The antagonistic effects of GSK1440115, but not GSK1562590, could be reversed by washout in rat isolated aorta. In ex vivo studies, GSK1562590 inhibited hU-II-induced contraction of rat aorta for at least 24 h following dosing. Dissociation of GSK1562590 binding was considerably slower at rat than monkey UT. CONCLUSIONS AND IMPLICATIONS Whereas both GSK1440115 and GSK1562590 represent high-affinity/selective UT antagonists suitable for assessing the (patho)physiological role of U-II, only GSK1562590 exhibited sustained UT residence time and improved preclinical efficacy in vivo.
Collapse
Affiliation(s)
- D J Behm
- Metabolic Pathways Center of Excellence for Drug Discovery, GlaxoSmithKline, King of Prussia, PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gao S, Oh YB, Shah A, Park WH, Chung MJ, Lee YH, Kim SH. Urotensin II receptor antagonist attenuates monocrotaline-induced cardiac hypertrophy in rats. Am J Physiol Heart Circ Physiol 2010; 299:H1782-9. [DOI: 10.1152/ajpheart.00438.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide with potent cardiovascular effects through a G protein-coupled receptor. Hypoxia stimulates the secretion of UII and atrial natriuretic peptide (ANP). However, the effect of UII on hypoxia-induced cardiac hypertrophy is still controversial. The present study was conducted to determine whether human UII (hUII)-mediated ANP secretion influences hypoxia-induced cardiac hypertrophy using in vitro and in vivo models. Hypoxia caused an increase in ANP secretion and a decrease in atrial contractility in isolated perfused beating rat atria. hUII (0.01 and 0.1 nM) attenuated hypoxia-induced ANP secretion without changing the atrial contractility, and the hUII effect was mediated by the UII receptor signaling involving phospholipase C, inositol 1,3,4 trisphosphate receptor, and protein kinase C. Rats treated with monocrotaline (MCT, 60 mg/kg) showed right ventricular hypertrophy with increases in pulmonary arterial pressure and its diameter and plasma levels of UII and ANP that were attenuated by the pretreatment with an UII receptor antagonist, urantide. An acute administration of hUII (5 μM injection plus 2.5 μM infusion for 15 min) decreased the plasma ANP level in MCT-treated rats but increased the plasma ANP level in MCT plus urantide-treated and sham-operated rats. These results suggest that hUII may deteriorate MCT-induced cardiac hypertrophy mainly through a vasoconstriction of the pulmonary artery and partly through the suppression of ANP secretion.
Collapse
Affiliation(s)
| | | | | | | | - Myoung Ja Chung
- Pathology, Diabetic Research Center, Chonbuk National University Medical School, Jeonju; and
| | - Young-Ho Lee
- Department of Physiology, College of Medicine, Yonsei University, Seoul, Korea
| | | |
Collapse
|
50
|
Woo KT, Lau YK, Zhao Y, Puong KY, Tan HB, Fook-Chong S, Wong KS, Chan CM. Urotensin 2 and Retinoic Acid Receptor Alpha (RARA) Gene Expression in IgA Nephropathy. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2010. [DOI: 10.47102/annals-acadmedsg.v39n9p705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Introduction: IgA nephropathy is a disease where the pathogenesis is still poorly understood. Deoxyribonucleic acid (DNA) microarray technique allows tens of thousands of gene expressions to be examined at the same time. Commercial availability of microarray genechips has made this powerful tool accessible for wider utilisation in the study of diseases. Materials and Methods: Seven patients with IgA nephropathy, 6 with minimal change nephrotic syndrome (MCNS) as patient controls and 7 normal healthy subjects were screened for the differential expression of genes, genome-wide. The Human Genome U133 Plus 2.0 Arrays (Affymetrix, USA) were used to quantitate the differential expression of 38,500 well-characterised human genes. Results: A total of 7761 gene expressions were identified that have an IgAN/Normal gene expression ratio of 0.06-fold to 5.58-fold. About 35% of the altered gene expressions have no gene title or just a hypothetical protein label such as FLJ30679. Most of the remaining 65% are identified proteins where their importance to IgAN is not immediately apparent at this time. Among the 30 most upregulated and 30 most downregulated genes are Urotensin 2 (upregulated 3.09-fold, P <0.05) and Fatty-acid binding protein 6 (downregulated to 0.12-fold, P <0.05). Retinoic acid receptor alpha (vitamin A receptor) was also found downregulated to 0.41-fold (P <0.005). Taqman real-time polymerase chain reaction (PCR) for urotensin 2 and retinoic acid receptor alpha (RARA) were performed on 20 patients with IgA nephropathy and 11 with Minimal Change Disease and the data correlated with various clinical indices. Conclusions: The findings suggest that there may be a therapeutic role for retinoic acid receptor alpha (RARA) in IgA nephropathy and a clinical monitoring role for Urotensin 2 in Minimal Change Disease.
Keywords: DNA microarray technology, Genome-wide gene expression
Collapse
Affiliation(s)
| | | | - Yi Zhao
- Singapore General Hospital, Singapore
| | | | | | | | | | | |
Collapse
|