1
|
Pedard M, Prevost L, Carpena C, Holleran B, Desrues L, Dubois M, Nicola C, Gruel R, Godefroy D, Deffieux T, Tanter M, Ali C, Leduc R, Prézeau L, Gandolfo P, Morin F, Wurtz O, Bonnard T, Vivien D, Castel H. The urotensin II receptor triggers an early meningeal response and a delayed macrophage-dependent vasospasm after subarachnoid hemorrhage in male mice. Nat Commun 2024; 15:8430. [PMID: 39341842 PMCID: PMC11439053 DOI: 10.1038/s41467-024-52654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Subarachnoid hemorrhage (SAH) can be associated with neurological deficits and has profound consequences for mortality and morbidity. Cerebral vasospasm (CVS) and delayed cerebral ischemia affect neurological outcomes in SAH patients, but their mechanisms are not fully understood, and effective treatments are limited. Here, we report that urotensin II receptor UT plays a pivotal role in both early events and delayed mechanisms following SAH in male mice. Few days post-SAH, UT expression is triggered by blood or hemoglobin in the leptomeningeal compartment. UT contributes to perimeningeal glia limitans astrocyte reactivity, microvascular alterations and neuroinflammation independent of CNS-associated macrophages (CAMs). Later, CAM-dependent vascular inflammation and subsequent CVS develop, leading to cognitive dysfunction. In an SAH model using humanized UTh+/h+ male mice, we show that post-SAH CVS and behavioral deficits, mediated by UT through Gq/PLC/Ca2+ signaling, are prevented by UT antagonists. These results highlight the potential of targeting UT pathways to reduce early meningeal response and delayed cerebral ischemia in SAH patients.
Collapse
Affiliation(s)
- Martin Pedard
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Lucie Prevost
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Camille Carpena
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Brian Holleran
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Laurence Desrues
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Martine Dubois
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Celeste Nicola
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Roxane Gruel
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - David Godefroy
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
- Univ Rouen Normandie, Inserm, Normandie Univ, NorDiC UMR 1239, Rouen, France
| | - Thomas Deffieux
- Institute Physics for Medicine, Inserm U1273, CNRS UMR 8631, ESPCI Paris, Paris Sciences et Lettres PSL University, Paris, France
| | - Mickael Tanter
- Institute Physics for Medicine, Inserm U1273, CNRS UMR 8631, ESPCI Paris, Paris Sciences et Lettres PSL University, Paris, France
| | - Carine Ali
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Laurent Prézeau
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Pierrick Gandolfo
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Fabrice Morin
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Olivier Wurtz
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Thomas Bonnard
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
- Centre Hospitalier Universitaire Caen, Department of Clinical Research, Caen, France
| | - Hélène Castel
- Univ Rouen Normandie, Inserm, Normandie Univ, CBG UMR 1245, Rouen, France.
- Institute of Research and Innovation in Biomedicine (IRIB), Rouen, France.
| |
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: 1] [Impact Index Per Article: 0.5] [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
|
The Role of Urotensin-II in Obesity and Metabolic Syndrome in Pediatric Population. CHILDREN 2022; 9:children9020204. [PMID: 35204924 PMCID: PMC8870523 DOI: 10.3390/children9020204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 12/04/2022]
Abstract
Background: Urotensin-II (U-II) is a short cyclic peptide that is widely recognized as one of the most potent vasoconstrictors. U-II plays a role in the pathophysiology of MS, participating in the development of essential hypertension, insulin resistance, hyperglycemia, and a proinflammatory state. Methods: This study comprised 52 obese children and adolescents with a body mass index (BMI) z score > 2, aged 10 to 18 years. Serum levels of U-II were assessed using an enzyme-linked immunosorbent assay along with other standard biochemical parameters. Results: Elevated serum levels of U-II were recorded in the group of obese subjects with MS when compared with the group of obese subjects without MS (4.99 (8.97–3.16) vs. 4.17 (5.17–2.03) ng/mL, median and IQR, p = 0.026). Furthermore, a subgroup of study subjects with high blood pressure had significantly higher U-II levels in comparison with the normotensive subgroup (4.98 (7.19–3.22) vs. 3.32 (5.06–1.97) ng/mL, p = 0.027), while the subgroup with a positive family history of high blood pressure had significantly higher U-II levels when compared with subjects who had a negative family history of elevated blood pressure (5.06 (6.83–4.45) vs. 3.32 (6.13–2.21) ng/mL, p = 0.039). Conclusions: To the best of the author’s knowledge, this is the first study on the levels of U-II in obese children and adolescents, including a possible link to MS.
Collapse
|
4
|
Lim G, Lim CJ, Lee JH, Lee BH, Ryu JY, Oh KS. Identification of new target proteins of a Urotensin-II receptor antagonist using transcriptome-based drug repositioning approach. Sci Rep 2021; 11:17138. [PMID: 34429474 PMCID: PMC8384862 DOI: 10.1038/s41598-021-96612-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/11/2021] [Indexed: 12/20/2022] Open
Abstract
Drug repositioning research using transcriptome data has recently attracted attention. In this study, we attempted to identify new target proteins of the urotensin-II receptor antagonist, KR-37524 (4-(3-bromo-4-(piperidin-4-yloxy)benzyl)-N-(3-(dimethylamino)phenyl)piperazine-1-carboxamide dihydrochloride), using a transcriptome-based drug repositioning approach. To do this, we obtained KR-37524-induced gene expression profile changes in four cell lines (A375, A549, MCF7, and PC3), and compared them with the approved drug-induced gene expression profile changes available in the LINCS L1000 database to identify approved drugs with similar gene expression profile changes. Here, the similarity between the two gene expression profile changes was calculated using the connectivity score. We then selected proteins that are known targets of the top three approved drugs with the highest connectivity score in each cell line (12 drugs in total) as potential targets of KR-37524. Seven potential target proteins were experimentally confirmed using an in vitro binding assay. Through this analysis, we identified that neurologically regulated serotonin transporter proteins are new target proteins of KR-37524. These results indicate that the transcriptome-based drug repositioning approach can be used to identify new target proteins of a given compound, and we provide a standalone software developed in this study that will serve as a useful tool for drug repositioning.
Collapse
Affiliation(s)
- Gyutae Lim
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Chae Jo Lim
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Jeong Hyun Lee
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Byung Ho Lee
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Jae Yong Ryu
- Department of Biotechnology, Duksung Women's University, 33 Samyang-ro 144-gil, Dobong-gu, Seoul, 01369, Republic of Korea.
| | - Kwang-Seok Oh
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea. .,Department of Medicinal and Pharmaceutical Chemistry, University of Science and Technology, 217 Gajeong-ro, Yuseong,-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
5
|
Dynamic Changes in Plasma Urotensin II and Its Correlation With Plaque Stability. J Cardiovasc Pharmacol 2021; 78:e147-e155. [PMID: 34173803 DOI: 10.1097/fjc.0000000000001044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/20/2021] [Indexed: 11/27/2022]
Abstract
ABSTRACT Urotensin II (UII) is involved in the formation of atherosclerosis, but its role in the stability of atherosclerotic plaques is unknown. The purpose of this study was to observe the dynamic changes in plasma UII and analyze its relationship to the stability of atherosclerotic plaques. One hundred thirty-five consecutive patients with acute coronary syndrome (ACS) were enrolled. The plasma UII levels were measured immediately after admission and during three-month follow-up. A vulnerable plaque model was established using local transfection of a recombinant P53 adenovirus into plaques in rabbits fed with a high-cholesterol diet and subjected to balloon arterial injury. The levels of plasma UII were measured weekly. The changes in plasma UII during the formation of atherosclerotic plaques and before and after plaque transfection were observed. The morphology of the plaques and the expression, distribution, and quantitative expression of UII in the plaques also were observed. Our results showed that the levels of plasma UII in patients with ACS at admission were lower than levels observed at the three-month follow-up. UII dynamic changes and its correlation with plaque stabilities were further verified in rabbits with atherosclerotic vulnerable plaques. The UII levels in rabbits were significantly decreased immediately after the P53 gene transfection, which led to plaque instability and rupture. These results suggested that UII expression was down-regulated in ACS, which may be related to its ability to modulate mechanisms involved in plaque stability and instability.
Collapse
|
6
|
Cui H, Lin Y, Xie L, Zhao J. Urantide decreases hepatic steatosis in rats with experimental atherosclerosis via the MAPK/Erk/JNK pathway. Mol Med Rep 2021; 23:284. [PMID: 33604686 PMCID: PMC7905324 DOI: 10.3892/mmr.2021.11923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
Hepatic steatosis, an indicator of atherosclerosis (AS), is always accompanied by inflammatory responses and disturbances in lipid metabolism. The present study investigated the protective effect of urantide, a urotensin II (UII) receptor antagonist, on the liver of rats with AS with hepatic steatosis by regulating the MAPK pathway. AS was induced in rats via an intraperitoneal injection of vitamin D3 and the administration of a high‑fat diet. Urantide treatment was then administered to the rats. Pathology, liver index, lipid levels and liver function were measured to determine liver injury. The expression levels of UII and G protein‑coupled receptor 14 (GPR14) were determined using immunohistochemistry, reverse transcription‑quantitative PCR and western blotting. The expression levels of MAPK‑related proteins in hepatocytes from each group were quantified using western blotting and immunofluorescence staining. Rats with AS had typical pathological changes associated with AS and hepatic steatosis, which were significantly improved by urantide treatment. Blood lipid levels, body weight, liver index and liver function were recovered in rats with AS after urantide treatment. Urantide downregulated the expression levels of UII and GPR14 in the livers of rats with AS; concurrently, the phosphorylation of Erk1/2 and JNK was significantly decreased. Moreover, no significant changes were observed in the phosphorylation of p38 MAPK in AS rat livers. In conclusion, urantide inhibits the activation of Erk1/2 and JNK by blocking the binding of UII and GPR14, thereby alleviating hepatic steatosis in rats with AS, ultimately restoring lipid metabolism in the liver and alleviating AS lesions.
Collapse
Affiliation(s)
- Haipeng Cui
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Yingxue Lin
- Department of Medicine, Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Lide Xie
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Juan Zhao
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
7
|
Konno N, Takano M, Miura K, Miyazato M, Nakamachi T, Matsuda K, Kaiya H. Identification and signaling characterization of four urotensin II receptor subtypes in the western clawed frog, Xenopus tropicalis. Gen Comp Endocrinol 2020; 299:113586. [PMID: 32828811 DOI: 10.1016/j.ygcen.2020.113586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/04/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
Urotensin II (UII) is involved, via the UII receptor (UTR), in many physiological and pathological processes, including vasoconstriction, locomotion, osmoregulation, immune response, and metabolic syndrome. In silico studies have revealed the presence of four or five distinct UTR (UTR1-UTR5) gene sequences in nonmammalian vertebrates. However, the functionality of these receptor subtypes and their associations to signaling pathways are unclear. In this study, full-length cDNAs encoding four distinct UTR subtypes (UTR1, UTR3, UTR4, and UTR5) were isolated from the western clawed frog (Xenopus tropicalis). In functional analyses, homologous Xenopus UII stimulation of cells expressing UTR1 or UTR5 induced intracellular calcoum mobilization and phosphorylation of extracellular signal-regulated kinase 1/2. Cells expressing UTR3 or UTR4 did not show this response. Furthermore, UII induced the phosphorylation of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) through the UII-UTR1/5 system. However, intracellular cAMP accumulation was not observed, suggesting that UII-induced CREB phosphorylation is caused by a signaling pathway different from that involving Gs protein. In contrast, the administration of UII to cells increased the phosphorylation of guanine nucleotide exchange factor-H1 (GEF-H1) and myosin light chain 2 (MLC2) in all UTR subtypes. These results define four distinct UTR functional subtypes and are consistent with the molecular evolution of UTR subtypes in vertebrates. Further understanding of signaling properties associated with UTR subtypes may help in clarifying the functional roles associated with UII-UTR interactions in nonmammalian vertebrates.
Collapse
Affiliation(s)
- Norifumi Konno
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan.
| | - Moe Takano
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Koichi Miura
- Department of Biochemistry, National Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, Osaka 564-8565, Japan; Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, Osaka 564-8565, Japan
| | - Tomoya Nakamachi
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Kouhei Matsuda
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, Osaka 564-8565, Japan
| |
Collapse
|
8
|
Watanabe T, Sato K. Roles of the kisspeptin/GPR54 system in pathomechanisms of atherosclerosis. Nutr Metab Cardiovasc Dis 2020; 30:889-895. [PMID: 32409274 DOI: 10.1016/j.numecd.2020.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/11/2019] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
AIMS Kisspeptin-10 (KP-10), a potent vasoconstrictor and inhibitor of angiogenesis, and its receptor, GPR54, have currently received much attention with respect to atherosclerosis, since both KP-10 and GPR54 are expressed at high levels in atheromatous plaques and restenotic lesions after wire-injury. The present review introduces the emerging roles of the KP-10/GPR54 system in atherosclerosis. DATA SYNTHESIS KP-10 suppresses migration and proliferation of human umbilical vein endothelial cells (HUVECs), and induces senescence in HUVECs. KP-10 increases adhesion of human monocytes to HUVECs. KP-10 also stimulates expression of interleukin-6, tumor necrosis factor-α, monocyte chemotactic protein-1, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin genes in HUVECs. KP-10 enhances oxidized low-density lipoprotein-induced foam cell formation associated with upregulation of CD36 and acyl-coenzyme A: cholesterol acyltransferase-1 in human monocyte-derived macrophages. In human aortic smooth muscle cells, KP-10 suppresses angiotensin II-induced migration and proliferation, however, it enhances apoptosis and activities of matrix metalloproteinase (MMP)-2 and MMP-9 by upregulation of extracellular signal-regulated kinase 1/2, p38, Bax, and caspase-3. Four-week-infusion of KP-10 into Apoe-/- mice accelerates development of aortic atherosclerotic lesions with increased monocyte/macrophage infiltration and vascular inflammation, also, it decreases intraplaque vascular smooth muscle cell content. Proatherosclerotic effects of endogenous and exogenous KP-10 were completely attenuated upon infusion of P234, a GPR54 antagonist, in Apoe-/- mice. CONCLUSION These findings suggest that KP-10 may contribute to acceleration of progression and to the instability of atheromatous plaques, leading to rupture of plaques. This GPR54 antagonist may be useful for the prevention and treatment of atherosclerosis. Thus, the KP-10/GPR54 system may serve as a novel therapeutic target for atherosclerotic diseases.
Collapse
Affiliation(s)
- Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; Department of Internal Medicine, Ushioda General Hospital/Clinic, Yokohama, Japan.
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan; Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
9
|
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: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/30/2019] [Indexed: 12/26/2022] Open
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
|
10
|
Pereira-Castro J, Brás-Silva C, Fontes-Sousa AP. Novel insights into the role of urotensin II in cardiovascular disease. Drug Discov Today 2019; 24:2170-2180. [PMID: 31430542 DOI: 10.1016/j.drudis.2019.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/26/2019] [Accepted: 08/12/2019] [Indexed: 12/16/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide that interacts with a specific receptor called the UT receptor. UII has been implicated in cardiovascular regulation, with promising therapeutic applications based on UT receptor antagonism. The endogenous ligands of the UT receptor: UII and urotensin-related peptide (URP), differentially bind and activate this receptor. Also, the receptor localization is not restricted to the plasma membrane, possibly inducing different physiological responses that could support its inconsistent, but potent, vasoactive activity. These properties could explain the disappointing outcomes in clinical studies, in contrast to the positive preclinical results regarding heart failure, pulmonary hypertension, atherosclerosis and diabetes mellitus. These aspects should be considered in future investigations to a better comprehension of the role of UII as a potential therapeutic target.
Collapse
Affiliation(s)
- João Pereira-Castro
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology, UnIC - Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ana Patrícia Fontes-Sousa
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal.
| |
Collapse
|
11
|
Lu D, Peng F, Li J, Zhao J, Ye X, Li B, Ding W. Urotensin II promotes secretion of LTB 4 through 5-lipoxygenase via the UT-ROS-Akt pathway in RAW264.7 macrophages. Arch Med Sci 2019; 15:1065-1072. [PMID: 31360201 PMCID: PMC6657259 DOI: 10.5114/aoms.2019.85197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/10/2017] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Urotensin II (UII) is an important vasoactive peptide involved in the pathogenesis of atherosclerosis. Monocytes/macrophages play important roles in every step of atherosclerosis. Although UII has a chemoattractant effect on monocytes, it is unclear whether UII regulates inflammatory responses in macrophages. The present study sought to explore whether UII can promote leukotriene B4 (LTB4) production by macrophages. MATERIAL AND METHODS The mRNA expression level of LTB4 and 5-lipoxygenase were determined by real-time polymerase chain reaction. The protein level of LTB4 and 5-lipoxygenase expression was assayed by enzyme-linked immunosorbent assay and Western blot, respectively. Western blot analysis was also employed to determine the phosphorylated forms of Akt. Reactive oxygen species (ROS) level was detected by the fluorescent probe 2',7'-dichlorofluorescin diacetate and fluorescence intensity was measured with a multiwell fluorescence plate reader. RESULTS Urotensin II promoted LTB4 release and increased 5-lipoxygenase expression in a concentration- and time-dependent manner in RAW264.7 cells. Leukotriene B4 production and 5-lipoxygenase expression were decreased by blocking the UII receptor (UT) with urantide, eliminating ROS with N-acetylcysteine and diphenyliodonium, and inhibiting Akt phosphorylation with LY294002. UII significantly elevated ROS production, whereas urantide, N-acetylcysteine and diphenyliodonium substantially attenuated this effect. UII also enhanced Akt phosphorylation significantly, and this effect was potently inhibited by urantide, N-acetylcysteine, diphenyliodonium and LY294002. CONCLUSIONS Urotensin II may promote 5-lipoxygenase expression and LTB4 release in RAW264.7 macrophages via UT-ROS-Akt pathways. These results indicate that UII may participate in macrophage activation and suggest a potential new mechanism underlying atherosclerosis.
Collapse
Affiliation(s)
- Dan Lu
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Fen Peng
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jun Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jing Zhao
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Xiaojin Ye
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Binghan Li
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Wenhui Ding
- Department of Cardiology, Peking University First Hospital, Beijing, China
| |
Collapse
|
12
|
Demirpence M, Guler A, Yilmaz H, Sayin A, Pekcevik Y, Turkon H, Colak A, Ari EM, Aslanipour B, Kocabas GU, Calan M. Is elevated urotensin II level a predictor for increased cardiovascular risk in subjects with acromegaly? J Endocrinol Invest 2019; 42:207-215. [PMID: 29804270 DOI: 10.1007/s40618-018-0905-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/18/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Acromegaly is a rare disorder existed in the result of overproduction of growth hormone (GH). The disorder is associated with increased cardiovascular risk factors and metabolic abnormalities. Urotensin II (UII), a secreted vasoactive peptide hormone, belonging somatostatin superfamily, plays an essential role in atherosclerosis and glucose metabolism. The aim of this study was to ascertain whether circulating UII levels are altered in subjects with acromegaly, and to describe the relationship between UII and hormonal or cardiometabolic parameters. METHODS This cross-sectional study included 41 subjects with active acromegaly, 28 subjects with controlled acromegaly, and 37 age- and BMI-matched controls without acromegaly. Hormonal and metabolic features of the subjects as well as carotid intima media thickness (cIMT) and epicardial fat thickness (EFT) were defined. Circulation of UII levels was determined via ELISA. RESULTS Both active and controlled acromegalic subjects showed a significant elevation of circulating levels of UII with respect to controls. There was no remarkable difference in circulating levels of UII between active and controlled acromegalic groups. Both cIMT and EFT were remarkably increased in acromegaly subjects comparing to controls. UII positively correlated with cIMT, EFT, BMI, and HOMA-IR. There was no correlation between UII and GH, insulin-like growth factor-1. According to the results obtained from regression models, UII levels independently predicted cIMT and EFT. CONCLUSION Elevated UII levels are associated with severity of cardiovascular risk factors including cIMT and EFT in acromegalic subjects.
Collapse
Affiliation(s)
- M Demirpence
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Izmir Tepecik Training and Research Hospital, Tepecik, 35120, Izmir, Turkey
| | - A Guler
- Department of Family Physician, Izmir Bozyaka Training and Research Hospital, Bozyaka, 35170, Izmir, Turkey
| | - H Yilmaz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Izmir Tepecik Training and Research Hospital, Tepecik, 35120, Izmir, Turkey
| | - A Sayin
- Department of Cardiology, Izmir Tepecik Research and Training Hospital, Tepecik, 35120, Izmir, Turkey
| | - Y Pekcevik
- Department of Radiology, Izmir Tepecik Training and Research Hospital, Tepecik, 35120, Izmir, Turkey
| | - H Turkon
- Department of Medical Biochemistry, Faculty of Medicine, Canakkale Onsekiz Mart University, 17120, Canakkale, Turkey
| | - A Colak
- Department of Clinical Biochemistry, Izmir Tepecik Training and Research Hospital, Tepecik, 35120, Izmir, Turkey
| | - E M Ari
- Department of Clinical Biochemistry, Izmir Tepecik Training and Research Hospital, Tepecik, 35120, Izmir, Turkey
| | - B Aslanipour
- Department of Bioengineering, Faculty of Engineering, Ege University, Bornova, 35100, Izmir, Turkey
| | - G U Kocabas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Izmir Bozyaka Training and Research Hospital, Bozyaka, 35170, Izmir, Turkey
| | - M Calan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Izmir Bozyaka Training and Research Hospital, Bozyaka, 35170, Izmir, Turkey.
| |
Collapse
|
13
|
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: 3.6] [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
|
14
|
Takahashi Y, Watanabe R, Sato Y, Ozawa N, Kojima M, Watanabe-Kominato K, Shirai R, Sato K, Hirano T, Watanabe T. Novel phytopeptide osmotin mimics preventive effects of adiponectin on vascular inflammation and atherosclerosis. Metabolism 2018; 83:128-138. [PMID: 29410350 DOI: 10.1016/j.metabol.2018.01.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/08/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The novel phytohormone, osmotin, has been reported to act like mammalian adiponectin through PHO36/AdipoR1 in various in vitro and in vivo models. However, there have been no reports regarding the precise effects of osmotin on atherosclerosis. METHODS We assessed the atheroprotective effects of osmotin on inflammatory molecules in human umbilical vein endothelial cells (HUVECs), human leukemic monocyte (THP-1) adhesion, inflammatory responses, and foam cell formation in THP-1-derived macrophages, and the migration, proliferation, and extracellular matrix expression in human aortic smooth muscle cells (HASMCs). We examined whether 4-week infusion of osmotin could suppress the development of aortic atherosclerotic lesions in apolipoprotein E-deficient (ApoE-/-) mice. RESULTS AdipoR1 was abundantly expressed in HUVECs, HASMCs, THP-1, and derived macrophages. Osmotin suppressed lipopolysaccharide-induced upregulation of tumor necrosis factor-α (TNF-α), monocyte chemotactic protein-1, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin in HUVECs, and TNF-α-induced THP-1-HUVEC adhesion. In THP-1-derived macrophages, osmotin suppressed the inflammatory M1 phenotype, lipopolysaccharide-induced secretion of interleukin-6 and TNF-α, and oxidized low-density lipoprotein-induced foam cell formation associated with CD36 and acyl-CoA:cholesterol acyltransferase 1 downregulation and ATP-binding cassette transporter A1 upregulation. In HASMCs, osmotin suppressed angiotensin II-induced migration, proliferation, collagen-1 and fibronectin expression, and matrix metalloproteinase-2 activity without inducing apoptosis. Infusion of osmotin into ApoE-/- mice prevented the development of aortic atherosclerotic lesions with reductions of intraplaque pentraxin-3 expression, fasting plasma glucose, and insulin resistance. CONCLUSIONS This study provided the first evidence that osmotin exerts preventive effects on vascular inflammation and atherosclerosis, which may facilitate the development of new therapeutic modalities for combating atherosclerosis and related diseases.
Collapse
Affiliation(s)
- Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yuki Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Nana Ozawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Miho Kojima
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Kaho Watanabe-Kominato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Remina Shirai
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan.
| |
Collapse
|
15
|
Rahimi R, Karimi J, Khodadadi I, Tayebinia H, Kheiripour N, Hashemnia M, Goli F. Silymarin ameliorates expression of urotensin II (U-II) and its receptor (UTR) and attenuates toxic oxidative stress in the heart of rats with type 2 diabetes. Biomed Pharmacother 2018; 101:244-250. [PMID: 29494961 DOI: 10.1016/j.biopha.2018.02.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/02/2018] [Accepted: 02/19/2018] [Indexed: 10/17/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with an increased risk of cardiovascular disease (CVD). Urotensin II ((U-II)) and its receptor (UTR) are involved in the progression of CVD through enhancement in the production of reactive oxygen species (ROS). Since silymarin (SMN) is a natural agent with anti-diabetic effects, this study aimed to investigate the antioxidant potency of SMN on the expression of (U-II)/UTR system and oxidative stress status in the heart of type 2 diabetic rats. Thirty-six male Wistar rats were randomly divided into six groups (n = 6). Control and diabetic groups treated with or without SMN (60 and 120 mg/kg/day) for 2 months. Fasting blood sugar (FBS), insulin, lipid profile, creatine kinase-MB ((CK-MB)), lactate dehydrogenase (LDH) and markers of oxidative stress were measured by spectrophotometric methods while (U-II) and UTR gene expression was determined by qPCR method. SMN significantly reduced the FBS level, increased the concentration of insulin and improved HOMA-IR. SMN prevented diabetes-induced weight loss, and attenuated the increased levels of total oxidative status (TOS), malondialdehyde (MDA), and nitric oxide (NO). Diabetes-induced reduction of total thiol molecules content (TTM) was normalized to the normal level in SMN treated rats. SMN significantly modulated serum lipid profile, reduced the expression of (U-II) and UTR in the heart, and improved histopathological changes in the heart tissues. Therefore, the current study indicated that SMN ameliorated unpleasant diabetic characteristics via down-regulation of (U-II) and UTR gene expression and modulation of oxidative stress in the heart tissue of type 2 diabetic rats.
Collapse
Affiliation(s)
- Rahimeh Rahimi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Iraj Khodadadi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Heidar Tayebinia
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nejat Kheiripour
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hashemnia
- Departments of Pathobiology, Veterinary Medicine Faculty, Razi University, Kermanshah, Iran
| | - Fatemeh Goli
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
16
|
Li J, Zhang YG, Luo LM, Dong X, Ding WH, Dang SY. Urotensin II promotes aldosterone expression in rat aortic adventitial fibroblasts. Mol Med Rep 2018; 17:2921-2928. [PMID: 29257277 PMCID: PMC5783511 DOI: 10.3892/mmr.2017.8233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 11/24/2017] [Indexed: 02/06/2023] Open
Abstract
Urotensin II (UII) contributes to cardiovascular diseases by activating vasoactive peptides. The present study aimed to determine the effect of UII on aldosterone (ALD) and its receptor in cultured adventitial fibroblasts (AFs) and the tunica adventitia of rat vessels to explore the possible mechanisms underlying vascular remodeling. Expression levels of aldosterone and its receptor on tunica adventitia were determined using immunohistochemistry. Growth‑arrested AFs and tunica adventitia from rat vessels were incubated with UII and inhibitors of various signal transduction pathways. ALD receptor (ALD‑R) mRNA expression levels and ALD protein exoression levels were determined by reverse transcription‑quantitative polymerase chain reaction and ELISA, respectively. Aldosterone and its receptors were expressed on tunica adventitia. UII promoted ALD protein secretion from cells in a dose‑ and time‑dependent manner. ALD‑R mRNA expression in cells was also dysregulated. Furthermore, the effects of UII were substantially inhibited by treatment with the inhibitors PD98059, Y‑27632, H‑7, CSA and nicardipine. These results were further verified in the tunica adventitia of rat vessels. The present findings indicated that UII stimulated ALD protein secretion and ALD‑R mRNA expression in AFs and in the tunica adventitia of rat vessels; moreover, this effect may be mediated by signal transduction pathways involving MAPK, Rho, PKC, calcineurin and Ca2+. UII may also contribute to vascular remodeling by stimulating the production of ALD and its receptor.
Collapse
Affiliation(s)
- Jun Li
- Division of Cardiology, Department of Internal Medicine, Peking University First Hospital, Beijing 100034, P.R. China
- Division of Cardiology, Department of Internal Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yong-Gang Zhang
- Department of Cardiovascular Diseases, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Li-Min Luo
- Department of Dermatology, Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiao Dong
- Division of Cardiology, Department of Internal Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wen-Hui Ding
- Division of Cardiology, Department of Internal Medicine, Peking University First Hospital, Beijing 100034, P.R. China
- Correspondence to: Professor Wen-Hui Ding, Division of Cardiology, Department of Internal Medicine, Peking University First Hospital, 8 Xishikudajie, Beijing 100034, P.R. China, E-mail:
| | - Shu-Yi Dang
- Division of Cardiology, Department of Internal Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
- Professor Shu-Yi Dang, Division of Cardiology, Department of Internal Medicine, Taihe Hospital, Hubei University of Medicine, 32 South People's Road, Shiyan, Hubei 442000, P.R. China, E-mail:
| |
Collapse
|
17
|
Terasaki M, Hiromura M, Mori Y, Kohashi K, Kushima H, Koshibu M, Saito T, Yashima H, Watanabe T, Hirano T. A Dipeptidyl Peptidase-4 Inhibitor Suppresses Macrophage Foam Cell Formation in Diabetic db/db Mice and Type 2 Diabetes Patients. Int J Endocrinol 2018; 2018:8458304. [PMID: 30627161 PMCID: PMC6304851 DOI: 10.1155/2018/8458304] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/04/2018] [Indexed: 11/24/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors could have antiatherosclerotic action, in addition to antihyperglycemic roles. Because macrophage foam cells are key components of atherosclerosis, we investigated the effect of the DPP-4 inhibitor teneligliptin on foam cell formation and its related gene expression levels in macrophages extracted from diabetic db/db (C57BLKS/J Iar -+Leprdb/+Leprdb ) mice and type 2 diabetes (T2D) patients ex vivo. We incubated mouse peritoneal macrophages and human monocyte-derived macrophages differentiated by 7-day culture with oxidized low-density lipoprotein in the presence/absence of teneligliptin (10 nmol/L) for 18 hours. We observed remarkable suppression of foam cell formation by teneligliptin treatment ex vivo in macrophages isolated from diabetic db/db mice (32%) and T2D patients (38%); this effect was accompanied by a reduction of CD36 (db/db mice, 43%; T2D patients, 46%) and acyl-coenzyme A: cholesterol acyltransferase-1 (ACAT-1) gene expression levels (db/db mice, 47%; T2D patients, 45%). Molecular mechanisms underlying this effect are associated with downregulation of CD36 and ACAT-1 by teneligliptin. The suppressive effect of a DPP-4 inhibitor on foam cell formation in T2D is conserved across species and is worth studying to elucidate its potential as an intervention for antiatherogenesis in T2D patients.
Collapse
Affiliation(s)
- Michishige Terasaki
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Munenori Hiromura
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Yusaku Mori
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Kyoko Kohashi
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Hideki Kushima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Masakazu Koshibu
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Tomomi Saito
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Hironori Yashima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji City, Tokyo, Japan
| | - Tsutomu Hirano
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Kim MY, Ilyosbek S, Lee BH, Yi KY, Jung YS. A novel urotensin II receptor antagonist, KR-36676, prevents ABCA1 repression via ERK/IL-1β pathway. Eur J Pharmacol 2017; 803:174-178. [DOI: 10.1016/j.ejphar.2017.03.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 01/08/2023]
|
19
|
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: 2.1] [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
|
20
|
Atheroprotective Effects of Tumor Necrosis Factor-Stimulated Gene-6. JACC Basic Transl Sci 2016; 1:494-509. [PMID: 30167534 PMCID: PMC6113406 DOI: 10.1016/j.jacbts.2016.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/13/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6), an anti-inflammatory protein, was shown to be localized in the neointima of injury-induced rat arteries. However, the modulatory effect of TSG-6 on atherogenesis has not yet been reported. We aimed to evaluate the atheroprotective effects of TSG-6 on human endothelial cells (HECs), human monocyte-derived macrophages (HMDMs), human aortic smooth muscle cells (HASMCs) in vitro, and aortic lesions in apolipoprotein E-deficient mice, along with expression levels of TSG-6 in coronary lesions and plasma from patients with coronary artery disease (CAD). TSG-6 was abundantly expressed in HECs, HMDMs, and HASMCs in vitro. TSG-6 significantly suppressed cell proliferation and lipopolysaccharide-induced up-regulation of monocyte chemotactic protein-1, intercellular adhesion molecule-1, and vascular adhesion molecule-1 in HECs. TSG-6 significantly suppressed inflammatory M1 phenotype and suppressed oxidized low-density lipoprotein-induced foam cell formation associated with down-regulation of CD36 and acyl-CoA:cholesterol acyltransferase-1 in HMDMs. In HASMCs, TSG-6 significantly suppressed migration and proliferation, but increased collagen-1 and -3 expressions. Four-week infusion of TSG-6 into apolipoprotein E-deficient mice significantly retarded the development of aortic atherosclerotic lesions with decreased vascular inflammation, monocyte/macrophage, and SMC contents and increased collagen fibers. In addition, it decreased peritoneal M1 macrophages with down-regulation of inflammatory molecules and lowered plasma total cholesterol levels. In patients with CAD, plasma TSG-6 levels were significantly increased, and TSG-6 was highly expressed in the fibrous cap within coronary atherosclerotic plaques. These results suggest that TSG-6 contributes to the prevention and stability of atherosclerotic plaques. Thus, TSG-6 may serve as a novel therapeutic target for CAD.
Collapse
Key Words
- ABCA1, ATP-binding cassette transporter A1
- ACAT1, acyl-CoA:cholesterol acyltransferase-1
- AngII, angiotensin II
- ApoE−/−, apolipoprotein E deficient
- CAD, coronary artery disease
- ECM, extracellular matrix
- HASMC, human aortic smooth muscle cell
- HMDM, human monocyte-derived macrophage
- HUVEC, human umbilical vein endothelial cell
- MMP, matrix metalloproteinase
- TIMP, tissue inhibitor of metalloproteinase
- TSG, tumor necrosis factor stimulated gene
- TSG-6
- VSMC, vascular smooth muscle cell
- atherosclerosis
- coronary artery disease
- endothelial cell
- macrophage
- oxLDL, oxidized low-density lipoprotein
- vascular smooth muscle cell
Collapse
|
21
|
Kim YA, Lee DG, Yi KY, Lee BH, Jung YS. Blockade of Urotensin II Receptor Prevents Vascular Dysfunction. Biomol Ther (Seoul) 2016; 24:523-8. [PMID: 27582556 PMCID: PMC5012878 DOI: 10.4062/biomolther.2015.142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 02/07/2023] Open
Abstract
Urotensin II (UII) is a potent vasoactive peptide and mitogenic agent to induce proliferation of various cells including vascular smooth muscle cells (VSMCs). In this study, we examined the effects of a novel UII receptor (UT) antagonist, KR-36676, on vasoconstriction of aorta and proliferation of aortic SMCs. In rat aorta, UII-induced vasoconstriction was significantly inhibited by KR-36676 in a concentration-dependent manner. In primary human aortic SMCs (hAoSMCs), UII-induced cell proliferation was significantly inhibited by KR-36676 in a concentration-dependent manner. In addition, KR-36676 decreased UII-induced phosphorylation of ERK, and UII-induced cell proliferation was also significantly inhibited by a known ERK inhibitor U0126. In mouse carotid ligation model, intimal thickening of carotid artery was dramatically suppressed by oral treatment with KR-36676 (30 mg/ kg/day) for 4 weeks compared to vehicle-treated group. From these results, it is indicated that KR-36676 suppress UII-induced proliferation of VSMCs at least partially through inhibition of ERK activation, and that it also attenuates UII-induced vasoconstriction and vascular neointima formation. Our study suggest that KR-36676 may be an attractive candidate for the pharmacological management of vascular dysfunction.
Collapse
Affiliation(s)
- Young-Ae Kim
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Dong Gil Lee
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Kyu Yang Yi
- Research Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Byung Ho Lee
- Research Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea.,Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
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: 1.9] [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
|
Yamamoto K, Tajima Y, Hasegawa A, Takahashi Y, Kojima M, Watanabe R, Sato K, Shichiri M, Watanabe T. Contrasting effects of stanniocalcin-related polypeptides on macrophage foam cell formation and vascular smooth muscle cell migration. Peptides 2016; 82:120-127. [PMID: 27346255 DOI: 10.1016/j.peptides.2016.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/20/2016] [Accepted: 06/20/2016] [Indexed: 02/06/2023]
Abstract
Stanniocalcin (STC) is a calcium- and phosphate-regulating hormone secreted by the corpuscles of Stannius, an endocrine gland of bony fish. Its human homologues, STC1 and STC2 showing 34% amino acid identity each other, are expressed in a variety of human tissues. To clarify their roles in atherosclerosis, we investigated the effects of their full-length proteins, STC1(18-247) and STC2(25-302), and STC2-derived fragment peptides, STC2(80-100) and STC2(85-99), on inflammatory responses in human umbilical vein endothelial cells (HUVECs), human macrophage foam cell formation, the migration and proliferation of human aortic smooth muscle cells (HASMCs) and the extracellular matrix expression. All these polypeptides suppressed lipopolysaccharide-induced expressions of interleukin-6, monocyte chemotactic protein-1, and intercellular adhesion molecule-1 in HUVECs. Oxidized low-density lipoprotein-induced foam cell formation was significantly decreased by STC1(18-247) and increased by STC2(80-100) and STC2(85-99), but not STC2(25-302), in human macrophages. Expression of acyl-CoA:cholesterol acyltransferase-1 (ACAT1) was significantly suppressed by STC1(18-247) but stimulated by STC2(80-100) and STC2(85-99). Expression of ATP-binding cassette transporter A1 was significantly stimulated by STC1(18-247). Neither STC1(18-247) nor STC2-derived peptides significantly affected CD36 expression in human macrophages or HASMC proliferation. STC2(80-100) and STC2(85-99) significantly increased HASMC migration, whereas STC1(18-247) significantly suppressed the angiotensin II-induced HASMC migration. Expressions of collagen-1, fibronectin, matrix metalloproteinase-2, and elastin were mostly unchanged with the exception of fibronectin up-regulation by STC2(80-100). Our results demonstrated the contrasting effects of STC1 and STC2-derived peptides on human macrophage foam cell formation associated with ACAT1 expression and on HASMC migration. Thus, STC-related polypeptides could serve as a novel therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Keigo Yamamoto
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yukie Tajima
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Akinori Hasegawa
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Miho Kojima
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Masayoshi Shichiri
- Department of Endocrinology, Diabetes and Metabolism, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
| |
Collapse
|
24
|
Naito C, Hashimoto M, Watanabe K, Shirai R, Takahashi Y, Kojima M, Watanabe R, Sato K, Iso Y, Matsuyama TA, Suzuki H, Ishibashi-Ueda H, Watanabe T. Facilitatory effects of fetuin-A on atherosclerosis. Atherosclerosis 2016; 246:344-51. [DOI: 10.1016/j.atherosclerosis.2016.01.037] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/11/2022]
|
25
|
Watanabe K, Watanabe R, Konii H, Shirai R, Sato K, Matsuyama TA, Ishibashi-Ueda H, Koba S, Kobayashi Y, Hirano T, Watanabe T. Counteractive effects of omentin-1 against atherogenesis†. Cardiovasc Res 2016; 110:118-28. [PMID: 26790473 DOI: 10.1093/cvr/cvw016] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 01/12/2016] [Indexed: 01/28/2023] Open
Abstract
AIMS Omentin-1, a novel adipocytokine expressed in visceral fat tissue, is negatively correlated with obesity, insulin resistance, and stable coronary artery disease (CAD). However, there have been no previous reports regarding the effects of omentin-1 on atherogenesis. METHODS AND RESULTS This study was performed to evaluate the atheroprotective effects of omentin-1 on human monocyte-derived macrophages, human aortic smooth muscle cells (HASMCs) in vitro, and aortic lesions in Apoe(-/-) mice in vivo. The histological expression of omentin-1 in coronary artery lesions and epicardial adipose tissues and its plasma levels were compared between CAD and non-CAD patients. Omentin-1 was abundantly expressed in human umbilical vein endothelial cells, macrophages, HASMCs, and human coronary artery SMCs in vitro. Omentin-1 promoted anti-inflammatory M2 phenotype during differentiation of human monocytes into macrophages. Omentin-1 suppressed oxidized low-density lipoprotein-induced foam cell formation associated with down-regulation of CD36, scavenger receptor class A, and acyl-CoA:cholesterol acyltransferase-1 and up-regulation of neutral cholesterol ester hydrolase in human macrophages. Omentin-1 suppressed angiotensin II-induced migration and platelet-derived growth factor-BB-induced proliferation, and collagen-1 and -3 expression in HASMCs. Four-week infusion of omentin-1 into Apoe(-/-) mice retarded the development of aortic atherosclerotic lesions with reduced contents of monocytes/macrophages, SMCs, and collagen fibres along with peritoneal M2-activated macrophages with inflammasome down-regulation and lowered plasma total cholesterol levels. Omentin-1 levels were markedly reduced in coronary endothelium and epicardial fat but increased in plasma and atheromatous plaques (macrophages/SMCs) in CAD patients compared with non-CAD patients. CONCLUSION This study provided the first evidence that omentin-1 may serve as a novel therapeutic target for atherosclerosis and CAD.
Collapse
Affiliation(s)
- Kaho Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0393, Japan
| | - Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0393, Japan
| | - Hanae Konii
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0393, Japan
| | - Remina Shirai
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0393, Japan
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0393, Japan
| | - Taka-Aki Matsuyama
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Youichi Kobayashi
- Division of Cardiology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji-City, Tokyo 192-0393, Japan
| |
Collapse
|
26
|
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: 0.9] [Reference Citation Analysis] [Abstract] [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
|
27
|
Palabiyik SS, Karakus E, Akpinar E, Halici Z, Bayir Y, Yayla M, Kose D. The Role of Urotensin Receptors in the Paracetamol-Induced Hepatotoxicity Model in Mice: Ameliorative Potential of Urotensin II Antagonist. Basic Clin Pharmacol Toxicol 2015; 118:150-9. [PMID: 26176337 DOI: 10.1111/bcpt.12447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/02/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Saziye S. Palabiyik
- Department of Pharmaceutical Toxicology; Faculty of Pharmacy; Ataturk University; Erzurum Turkey
| | - Emre Karakus
- Department of Pharmacology and Toxicology; Faculty of Veterinary Medicine; Ataturk University; Erzurum Turkey
| | - Erol Akpinar
- Department of Pharmacology; Faculty of Medicine; Ataturk University; Erzurum Turkey
| | - Zekai Halici
- Department of Pharmacology; Faculty of Medicine; Ataturk University; Erzurum Turkey
| | - Yasin Bayir
- Department of Biochemistry; Faculty of Pharmacy; Ataturk University; Erzurum Turkey
| | - Muhammed Yayla
- Department of Pharmacology; Faculty of Medicine; Ataturk University; Erzurum Turkey
| | - Duygu Kose
- Department of Pharmacology; Faculty of Medicine; Ataturk University; Erzurum Turkey
| |
Collapse
|
28
|
Vaudry H, Leprince J, Chatenet D, Fournier A, Lambert DG, Le Mével JC, Ohlstein EH, Schwertani A, Tostivint H, Vaudry D. International Union of Basic and Clinical Pharmacology. XCII. Urotensin II, urotensin II-related peptide, and their receptor: from structure to function. Pharmacol Rev 2015; 67:214-58. [PMID: 25535277 DOI: 10.1124/pr.114.009480] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Urotensin II (UII) is a cyclic neuropeptide that was first isolated from the urophysis of teleost fish on the basis of its ability to contract the hindgut. Subsequently, UII was characterized in tetrapods including humans. Phylogenetic studies and synteny analysis indicate that UII and its paralogous peptide urotensin II-related peptide (URP) belong to the somatostatin/cortistatin superfamily. In mammals, the UII and URP genes are primarily expressed in cholinergic neurons of the brainstem and spinal cord. UII and URP mRNAs are also present in various organs notably in the cardiovascular, renal, and endocrine systems. UII and URP activate a common G protein-coupled receptor, called UT, that exhibits relatively high sequence identity with somatostatin, opioid, and galanin receptors. The UT gene is widely expressed in the central nervous system (CNS) and in peripheral tissues including the retina, heart, vascular bed, lung, kidney, adrenal medulla, and skeletal muscle. Structure-activity relationship studies and NMR conformational analysis have led to the rational design of a number of peptidic and nonpeptidic UT agonists and antagonists. Consistent with the wide distribution of UT, UII has now been shown to exert a large array of biologic activities, in particular in the CNS, the cardiovascular system, and the kidney. Here, we review the current knowledge concerning the pleiotropic actions of UII and discusses the possible use of antagonists for future therapeutic applications.
Collapse
Affiliation(s)
- Hubert Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jérôme Leprince
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Chatenet
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Alain Fournier
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David G Lambert
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Jean-Claude Le Mével
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Eliot H Ohlstein
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Adel Schwertani
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - Hervé Tostivint
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| | - David Vaudry
- Institut National de la Santé et de la Recherche Médicale, U982, Institute for Research and Innovation in Biomedicine, Mont-Saint-Aignan, France (H.V., J.L., D.V.), University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.V.); Institut National de la Recherche Scientifique-Institut Armand Frappier, Laval, Québec, Canada (D.C., A.F.); International Associated Laboratory Samuel de Champlain, University of Rouen, Mont-Saint-Aignan, France (H.V., J.L., D.C., A.F., D.V.); Department of Cardiovascular Sciences, Division of Anaesthesia, Critical Care and Pain Management, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester, United Kingdom (D.G.L.); Institut National de la Santé et de la Recherche Médicale, U1101, Laboratoire de Traitement de l'Information Médicale, Laboratoire de Neurophysiologie, Université Européenne de Bretagne, Brest, France (J.-C.L.M.); AltheRx Pharmaceuticals, Malvern, Pennsylvania (E.H.O.); Division of Cardiology, Montreal General Hospital, McGill University Health Center, Montreal, Québec, Canada (A.S.); and Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7221, Evolution des Régulations Endocriniennes, Muséum National d'Histoire Naturelle, Paris, France (H.T.)
| |
Collapse
|
29
|
Tomiyama S, Nakamachi T, Uchiyama M, Matsuda K, Konno N. Urotensin II upregulates migration and cytokine gene expression in leukocytes of the African clawed frog, Xenopus laevis. Gen Comp Endocrinol 2015; 216:54-63. [PMID: 25907658 DOI: 10.1016/j.ygcen.2015.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 03/24/2015] [Accepted: 04/12/2015] [Indexed: 01/05/2023]
Abstract
Urotensin II (UII) exhibits diverse physiological actions including vasoconstriction, locomotor activity, osmoregulation, and immune response via the UII receptor (UTR) in mammals. However, in amphibians the function of the UII-UTR system remains unknown. In the present study, we investigated the potential immune function of UII using leukocytes isolated from the African clawed frog, Xenopus laevis. Stimulation of male frogs with lipopolysaccharide increased mRNA expression of UII and UTR in leukocytes, suggesting that inflammatory stimuli induce activation of the UII-UTR system. Migration assays showed that both UII and UII-related peptide enhanced migration of leukocytes in a dose-dependent manner, and that UII effect was inhibited by the UTR antagonist urantide. Inhibition of Rho kinase with Y-27632 abolished UII-induced migration, suggesting that it depends on the activation of RhoA/Rho kinase. Treatment of isolated leukocytes with UII increased the expression of several cytokine genes including tumor necrosis factor-α, interleukin-1β, and macrophage migration inhibitory factor, and the effects were abolished by urantide. These results suggest that in amphibian leukocytes the UII-UTR system is involved in the activation of leukocyte migration and cytokine gene expression in response to inflammatory stimuli.
Collapse
Affiliation(s)
- Shiori Tomiyama
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Tomoya Nakamachi
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Minoru Uchiyama
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Kouhei Matsuda
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Norifumi Konno
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan.
| |
Collapse
|
30
|
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: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
- * E-mail:
| | - 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
|
31
|
Vasoprotective effects of urocortin 1 against atherosclerosis in vitro and in vivo. PLoS One 2014; 9:e110866. [PMID: 25462164 PMCID: PMC4251828 DOI: 10.1371/journal.pone.0110866] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/18/2014] [Indexed: 11/19/2022] Open
Abstract
AIM Atherosclerosis is the complex lesion that consists of endothelial inflammation, macrophage foam cell formation, vascular smooth muscle cell (VSMC) migration and proliferation, and extracellular matrix production. Human urocortin 1 (Ucn1), a 40-amino acid peptide member of the corticotrophin-releasing factor/urotensin I family, has potent cardiovascular protective effects. This peptide induces potent and long-lasting hypotension and coronary vasodilation. However, the relationship of Ucn1 with atherosclerosis remains unclear. The present study was performed to clarify the effects of Ucn1 on atherosclerosis. METHODS We assessed the effects of Ucn1 on the inflammatory response and proliferation of human endothelial cells (ECs), human macrophage foam cell formation, migration and proliferation of human VSMCs, extracellular matrix expression in VSMCs, and the development of atherosclerosis in apolipoprotein E-deficient (Apoe-/-) mice. RESULTS Ucn1 significantly suppressed cell proliferation without inducing apoptosis, and lipopolysaccharide-induced up-regulation of monocyte chemoattractant protein-1 and intercellular adhesion molecule-1 in human ECs. Ucn1 significantly reduced oxidized low-density lipoprotein-induced foam cell formation with a significant down-regulation of CD36 and acyl-CoA:cholesterol acyltransferase 1 in human monocyte-derived macrophages. Ucn1 significantly suppressed the migration and proliferation of human VSMCs and increased the activities of matrix metalloproteinase-2 (MMP2) and MMP9 in human VSMCs. Intraperitoneal injection of Ucn1 into Apoe-/- mice for 4 weeks significantly retarded the development of aortic atherosclerotic lesions. CONCLUSIONS This study provided the first evidence that Ucn1 prevents the development of atherosclerosis by suppressing EC inflammatory response and proliferation, macrophage foam cell formation, and VSMC migration and proliferation. Thus, Ucn1 could serve as a novel therapeutic target for atherosclerotic cardiovascular diseases.
Collapse
|
32
|
Wang Y, Wu JF, Tang YY, Zhang M, Li Y, Chen K, Zeng MY, Yao F, Xie W, Zheng XL, Zeng GF, Tang CK. Urotensin II increases foam cell formation by repressing ABCA1 expression through the ERK/NF-κB pathway in THP-1 macrophages. Biochem Biophys Res Commun 2014; 452:998-1003. [DOI: 10.1016/j.bbrc.2014.09.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 12/21/2022]
|
33
|
Al Kindi H, Hafiane A, You Z, Albanese I, Pilote L, Genest J, Schwertani A. Circulating levels of the vasoactive peptide urotensin II in patients with acute coronary syndrome and stable coronary artery disease. Peptides 2014; 55:151-7. [PMID: 24642358 DOI: 10.1016/j.peptides.2014.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/26/2014] [Accepted: 03/09/2014] [Indexed: 12/26/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide with various roles in cardiovascular physiology and pathophysiology. There is an accumulating evidence implicating UII in atherosclerosis and coronary artery disease, making it an important target in acute coronary syndrome (ACS). In this study, we sought to determine the plasma levels of UII in ACS patients within 48 h of clinical presentation and after a 12-week recovery period. We compared them to patients with stable coronary artery disease (CAD) and a control group of normolipidemic subjects without known CAD. Using a highly sensitive ELISA technique, we measured plasma UII in 27 ACS patients, 26 stable CAD patients and 22 age-matched controls. ACS patients had significantly elevated plasma UII during the first 48 h of clinical presentation compared to stable CAD patients and controls. We also found significant positive correlations between UII and CRP and with triglycerides and a significant negative correlation between UII and EF. There was no correlation with LDL-C. In conclusion, plasma UII levels were elevated in patients with acute coronary syndrome, particularly immediately after clinical presentation. This suggests an upregulation of UII expression in ACS.
Collapse
Affiliation(s)
- Hamood Al Kindi
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Anouar Hafiane
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Zhipeng You
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Isabella Albanese
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Louise Pilote
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jacques Genest
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Adel Schwertani
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
34
|
Li Y, Zhao S, Wang Y, Chen Y, Lin Y, Zhu N, Zheng H, Wu M, Cheng D, Li Y, Bai L, Fan J, Liu E. Urotensin II promotes atherosclerosis in cholesterol-fed rabbits. PLoS One 2014; 9:e95089. [PMID: 24747943 PMCID: PMC3991611 DOI: 10.1371/journal.pone.0095089] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/22/2014] [Indexed: 12/16/2022] Open
Abstract
Urotensin II (UII) is a vasoactive peptide composed of 11 amino acids that has been implicated to contribute to the development of cardiovascular disease. The purpose of this study was to investigate whether UII affects the development of atherosclerosis in cholesterol-fed rabbits. UII was infused for 16 weeks through an osmotic mini-pump into male Japanese White rabbits fed on a high-cholesterol diet. Plasma lipids and body weight were measured every 4 weeks. Aortic atherosclerotic lesions along with cellular components, collagen fibers, matrix metalloproteinase-1 and -9 were examined. Moreover, vulnerability index of atherosclerotic plaques was evaluated. UII infusion significantly increased atherosclerotic lesions within the entire aorta by 21% over the control (P = 0.013). Atherosclerotic lesions were increased by 24% in the aortic arch (P = 0.005), 11% in the thoracic aorta (P = 0.054) and 18% in the abdominal aorta (P = 0.035). These increases occurred without changes in plasma levels of total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglycerides or body weight. Immunohistochemical staining revealed that macrophages and matrix metalloproteinase-9 were significantly enhanced by 2.2-fold and 1.6-fold in UII group. In vitro studies demonstrated that UII up-regulated the expression of vascular cell adhesion protein-1 and intercellular adhesion molecule-1 in human umbilical vein endothelial cells, which was inhibited by the UII receptor antagonist urantide. In conclusion, our results showed that UII promotes the development of atherosclerotic lesions and destabilizes atherosclerotic plaques in cholesterol-fed rabbits.
Collapse
Affiliation(s)
- Yafeng Li
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Sihai Zhao
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Yanli Wang
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Yulong Chen
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Yan Lin
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Ninghong Zhu
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Huadong Zheng
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China
| | - Min Wu
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China
| | - Daxing Cheng
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China
| | - Yandong Li
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Liang Bai
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Enqi Liu
- Laboratory for Lipid Metabolism and Atherosclerosis, Xi'an Jiaotong University Cardiovascular Research Center, Xi'an, Shaanxi, China; Laboratory Animal Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
35
|
Tashiro Y, Sato K, Watanabe T, Nohtomi K, Terasaki M, Nagashima M, Hirano T. A glucagon-like peptide-1 analog liraglutide suppresses macrophage foam cell formation and atherosclerosis. Peptides 2014; 54:19-26. [PMID: 24418070 DOI: 10.1016/j.peptides.2013.12.015] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 12/27/2022]
Abstract
Macrophage foam cell formation, characterized by cholesterol ester accumulation catalyzed by acyl-CoA:cholesterol acyltransferase 1 (ACAT1), is the hallmark of early atherogenesis. We previously demonstrated the suppressive effects of incretins, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), on the development of atherosclerotic lesions in apolipoprotein E-deficient (apoE(-/-)) mice. The present study was performed to evaluate the suppressive effects of these incretins and GLP-1 analogs, such as exendin-4 and liraglutide, on human macrophage foam cell formation in vitro and those of liraglutide on atherosclerotic lesion development in apoE(-/-) mice. We investigated the suppressive effects of GLP-1, GIP, exendin-4, and liraglutide against oxidized low-density lipoprotein (oxLDL)-induced foam cell formation in primary cultured human monocyte-derived macrophages. Seventeen-week-old apoE(-/-) mice were administered a long-acting GLP-1 analog liraglutide by osmotic mini-pumps for 4 weeks. Aortic atherosclerosis, oxLDL-induced foam cell formation, and related gene expression in exudate peritoneal macrophages were determined in vivo and ex vivo. Receptors for GLP-1 and GIP were expressed at high levels in human aortic smooth muscle cells and monocytes, but at relatively low levels in human macrophages and foam cells. GLP-1, GIP, exendin-4, and liraglutide significantly suppressed oxLDL-induced foam cell formation mainly associated with ACAT1 down-regulation in human monocyte-derived macrophages. The infusion of liraglutide into apoE(-/-) mice significantly retarded atherosclerotic lesions with monocyte/macrophage infiltration in the aortic wall and suppressed foam cell formation and ACAT1 expression in macrophages. These findings indicate that liraglutide could prevent the development of atherosclerotic lesions by suppressing macrophage foam cell formation mainly associated with ACAT1 down-regulation.
Collapse
Affiliation(s)
- Yuko Tashiro
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
| | - Kyoko Nohtomi
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Michishige Terasaki
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Masaharu Nagashima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Tsutomu Hirano
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Mohammadi A, Najar AG, Khoshi A. Effect of urotensin II on apolipoprotein B100 and apolipoprotein A-I expression in HepG2 cell line. Adv Biomed Res 2014; 3:22. [PMID: 24600602 PMCID: PMC3929015 DOI: 10.4103/2277-9175.124661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/19/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Increased apolipoprotein B100 (apo B) and decreased apolipoprotein A-I (apo A-I) production are important risk factors in atherosclerosis. Urotensin II (UII), as the most potent vasoconstrictor in human, is related with hypertension and probably atherosclerosis. Because of the relationship between the hypertension and lipoprotein metabolism in atherosclerosis, the aim of this study was to test the effect of urotensin II on apo B and apo A-I expression in hepatic (HepG2) cell line. MATERIALS AND METHODS HepG2 cells were treated with 10, 50, 100, and 200 nmol/L of urotensin II (n = 6). Relative apo B and apo A-I messenger RNA (mRNA) levels in conditioned media, normalized to glyceraldehyde-3-phosphate dehydrogenase, were measured with quantitative real-time polymerase chain reaction method. In addition, apo B and apo A-I levels were also estimated and compared with the controls using the western blotting method. Data were analyzed statistically by ANOVA and non-parametric tests. RESULTS The apo B mRNA levels were not increased significantly following the treatment with UII. However, apo B protein levels were increased significantly after the treatment with urotensin II, especially at 100 and 200 nmol/L. The apo A-I mRNA and protein levels in conditioned media also were not significantly changed. However, there was a significant decrease in apo A-I mRNA and protein levels at 200 nM UII. CONCLUSIONS UII might increase apo B at protein level probably through participating factors in its synthesis and/ or stability/degradation. In addition, UII may have decreasing effect at more than 200 nM concentrations on apo A-I.
Collapse
Affiliation(s)
- Abbas Mohammadi
- Department of Biochemistry, Physiology Research Center, Kerman, Iran
| | | | - Amirhosein Khoshi
- Department of Biochemistry, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
37
|
Konii H, Sato K, Kikuchi S, Okiyama H, Watanabe R, Hasegawa A, Yamamoto K, Itoh F, Hirano T, Watanabe T. Stimulatory Effects of Cardiotrophin 1 on Atherosclerosis. Hypertension 2013; 62:942-50. [DOI: 10.1161/hypertensionaha.113.01653] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiotrophin 1 (CT-1), an interleukin-6 family cytokine, was recently shown to be expressed in the intima of early atherosclerotic lesions in the human carotid artery. CT-1 stimulates proatherogenic molecule expression in human vascular endothelial cells and monocyte migration. However, it has not been reported whether CT-1 accelerates atherosclerosis. This study was performed to examine the stimulatory effects of CT-1 on human macrophage foam cell formation and vascular smooth muscle cell migration and proliferation in vitro, and on the development of atherosclerotic lesions in apolipoprotein E–deficient (ApoE
−/−
) mice in vivo. CT-1 was expressed at high levels in endothelial cells and macrophages in both humans and ApoE
−/−
mice. CT-1 significantly enhanced oxidized low-density lipoprotein–induced foam cell formation associated with increased levels of CD36 and acyl-CoA:cholesterol acyltransferase-1 expression in human monocyte–derived macrophages. CT-1 significantly stimulated the migration, proliferation, and collagen-1 expression in human aortic vascular smooth muscle cells. Four-week infusion of CT-1 into ApoE
−/−
mice significantly accelerated the development of aortic atherosclerotic lesions with increased monocyte/macrophage infiltration, vascular smooth muscle cell proliferation, and collagen-1 content in the aortic wall. Activation of inflammasome, such as apoptosis-associated speck-like protein containing a caspase recruitment domain, nuclear factor κB, and cyclooxygenase-2, was observed in exudate peritoneal macrophages from ApoE
−/−
mice infused with CT-1. Infusion of anti–CT-1–neutralizing antibody alone into ApoE
−/−
mice significantly suppressed monocyte/macrophage infiltration in atherosclerotic lesions. These results indicate that CT-1 accelerates the development of atherosclerotic lesions by stimulating the inflammasome, foam cell formation associated with CD36 and acyl-CoA:cholesterol acyltransferase-1 upregulation in macrophages, and migration, proliferation, and collagen-1 production in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Hanae Konii
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Kengo Sato
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Sayaka Kikuchi
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Hazuki Okiyama
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Rena Watanabe
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Akinori Hasegawa
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Keigo Yamamoto
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Fumiko Itoh
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Tsutomu Hirano
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| | - Takuya Watanabe
- From the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Japan (H.K., K.S., S.K., H.O., R.W., A.H., K.Y., F.I., T.W.); and Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan (T.H.)
| |
Collapse
|
38
|
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]
|
39
|
Salusins: potential use as a biomarker for atherosclerotic cardiovascular diseases. Int J Hypertens 2013; 2013:965140. [PMID: 24251033 PMCID: PMC3819761 DOI: 10.1155/2013/965140] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/04/2013] [Accepted: 09/19/2013] [Indexed: 12/31/2022] Open
Abstract
Human salusin- α and salusin- β are related peptides produced from prosalusin. Bolus injection of salusin- β into rats induces more profound hypotension and bradycardia than salusin- α . Central administration of salusin- β increases blood pressure via release of norepinephrine and arginine-vasopressin. Circulating levels of salusin- α and salusin- β are lower in patients with essential hypertension. Salusin- β exerts more potent mitogenic effects on human vascular smooth muscle cells (VSMCs) and fibroblasts than salusin- α . Salusin- β accelerates inflammatory responses in human endothelial cells and monocyte-endothelial adhesion. Human macrophage foam cell formation is stimulated by salusin- β but suppressed by salusin- α . Chronic salusin- β infusion into apolipoprotein E-deficient mice enhances atherosclerotic lesions; salusin- α infusion reduces lesions. Salusin- β is expressed in proliferative neointimal lesions of porcine coronary arteries after stenting. Salusin- α and salusin- β immunoreactivity have been detected in human coronary atherosclerotic plaques, with dominance of salusin- β in macrophage foam cells, VSMCs, and fibroblasts. Circulating salusin- β levels increase and salusin- α levels decrease in patients with coronary artery disease. These findings suggest that salusin- β and salusin- α may contribute to proatherogenesis and antiatherogenesis, respectively. Increased salusin- β and/or decreased salusin- α levels in circulating blood and vascular tissue are closely linked with atherosclerosis. Salusin- α and salusin- β could be candidate biomarkers and therapeutic targets for atherosclerotic cardiovascular diseases.
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Urotensin II (UTS2), the most potent vasoconstrictor identified thus far, is an undecapeptide hormone with a structure that is highly conserved through mammalian phylogeny. In spite of its broad expression across the invertebrate and vertebrate world, the precise role of UTS2 in physiology and disease is still unknown. The first description of human UTS2 and its receptor brought initial promise of a potential therapeutic target for progressive renal disease, with vasoconstrictive and profibrotic actions within an autocrine and paracrine system and local renal generation that was upregulated with renal pathology. RECENT FINDINGS However, the last decade has not brought the successful development of new treatments first hoped for, with one small human clinical trial bearing negative results. What has become apparent is that the spectrum of actions of UTS2 is broad and often paradoxical. This ancient hormone has both vasoconstrictor and vasodilatory actions, has both profibrotic and antiapoptotic activity, as well as actions which are highly contextual on the particular vascular bed studied and on the presence or absence of superimposed disease state. SUMMARY With current development of newer UTS2 antagonists attempting to more closely replicate the ligand-receptor kinetics of UTS2 and its receptor, the focus on potential clinical applications of UTS2 inhibition has moved away from the kidney to the treatment of chronic lung and cardiovascular diseases.
Collapse
|
41
|
Konno N, Fujii Y, Imae H, Kaiya H, Mukuda T, Miyazato M, Matsuda K, Uchiyama M. Urotensin II receptor (UTR) exists in hyaline chondrocytes: a study of peripheral distribution of UTR in the African clawed frog, Xenopus laevis. Gen Comp Endocrinol 2013; 185:44-56. [PMID: 23399967 DOI: 10.1016/j.ygcen.2013.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/15/2013] [Accepted: 01/20/2013] [Indexed: 02/07/2023]
Abstract
Urotensin II (UII) and UII-related peptide (URP) exhibit diverse physiological actions including vasoconstriction, locomotor activity, osmoregulation, and immune response through UII receptor (UTR), which is expressed in the central nervous system and peripheral tissues of fish and mammals. In amphibians, only UII has been identified. As the first step toward elucidating the actions of UII and URP in amphibians, we cloned and characterized URP and UTR from the African clawed frog Xenopus laevis. Functional analysis showed that treatment of UII or URP with Chinese hamster ovary cells transfected with the cloned receptor increased the intracellular calcium concentration in a concentration-dependent manner, whereas the administration of the UTR antagonist urantide inhibited UII- or URP-induced Ca(2+) mobilization. An immunohistochemical study showed that UTR was expressed in the splenocytes and leukocytes isolated from peripheral blood, suggesting that UII and URP are involved in the regulation of the immune system. UTR was also localized in the apical membrane of the distal tubule of the kidney and in the transitional epithelial cells of the urinary bladder. This result supports the view that the UII/URP-UTR system plays an important role in osmoregulation of amphibians. Interestingly, immunopositive labeling for UTR was first detected in the chondrocytes of various hyaline cartilages (the lung septa, interphalangeal joint and sternum). The expression of UTR was also observed in the costal cartilage, tracheal cartilages, and xiphoid process of the rat. These novel findings probably suggest that UII and URP mediate the formation of the cartilaginous matrix.
Collapse
Affiliation(s)
- Norifumi Konno
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Watson AMD, Olukman M, Koulis C, Tu Y, Samijono D, Yuen D, Lee C, Behm DJ, Cooper ME, Jandeleit-Dahm KAM, Calkin AC, Allen TJ. Urotensin II receptor antagonism confers vasoprotective effects in diabetes associated atherosclerosis: studies in humans and in a mouse model of diabetes. Diabetologia 2013; 56:1155-65. [PMID: 23344731 DOI: 10.1007/s00125-013-2837-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS The small, highly conserved vasoactive peptide urotensin II (UII) is upregulated in atherosclerosis. However, its effects in diabetes-associated atherosclerosis have not been assessed. METHODS Endothelial cells were grown in normal- and high-glucose (5 and 25 mmol/l) media with and without UII (10⁻⁸ mol/l) and/or the UII receptor antagonist, SB-657510 (10⁻⁸ mol/l). Apoe knockout (KO) mice with or without streptozotocin-induced diabetes were treated with or without SB-657510 (30 mg kg⁻¹ day⁻¹; n = 20 per group) and followed for 20 weeks. Carotid endarterectomy specimens from diabetic and non-diabetic humans were also evaluated. RESULTS In high (but not normal) glucose medium, UII significantly increased CCL2 (encodes macrophage chemoattractant protein 1 [MCP-1]) gene expression (human aortic endothelial cells) and increased monocyte adhesion (HUVECs). UII receptor antagonism in diabetic Apoe KO mice significantly attenuated diabetes-associated atherosclerosis and aortic staining for MCP-1, F4/80 (macrophage marker), cyclooxygenase-2, nitrotyrosine and UII. UII staining was significantly increased in carotid endarterectomies from diabetic compared with non-diabetic individuals, as was staining for MCP-1. CONCLUSIONS/INTERPRETATION This is the first report to demonstrate that UII is increased in diabetes-associated atherosclerosis in humans and rodents. Diabetes-associated plaque development was attenuated by UII receptor antagonism in the experimental setting. Thus UII may represent a novel therapeutic target in the treatment of diabetes-associated atherosclerosis.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/immunology
- Aorta/metabolism
- Aorta/pathology
- Atherosclerosis/complications
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Cell Adhesion/drug effects
- Cells, Cultured
- Crosses, Genetic
- Diabetes Mellitus, Type 1/complications
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/prevention & control
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/immunology
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/pathology
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/immunology
- Pilot Projects
- Protective Agents/pharmacology
- Protective Agents/therapeutic use
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/metabolism
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Urotensins/antagonists & inhibitors
- Urotensins/biosynthesis
- Urotensins/metabolism
Collapse
Affiliation(s)
- A M D Watson
- Baker IDI Heart and Diabetes Research Institute, PO Box 6492 St Kilda Road Central, Melbourne, VIC 8008, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Guo XH, Feng ZJ. Role of urotensin-Ⅱ in the pathogenesis of liver cirrhosis and portal hypertension and collateral circulation. Shijie Huaren Xiaohua Zazhi 2012; 20:3536-3541. [DOI: 10.11569/wcjd.v20.i35.3536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Urotensin-Ⅱ (U-Ⅱ) is a somatostatin-like cyclic peptide which has a potent vasoactive effect and can promote vascular reconstruction and hyperplasia. Research shows that UⅡ plays an important role in the development of liver cirrhosis and portal hypertension. UⅡ influences intrahepatic resistance and splanchnic hemodynamics through a variety of pathways, causing portal hypertension and participating in the formation of esophageal and gastric varices. UⅡ receptor antagonists can reduce portal pressure in cirrhotic rats, but this finding need to be confirmed clinically.
Collapse
|
44
|
You Z, Genest J, Barrette PO, Hafiane A, Behm DJ, D'Orleans-Juste P, Schwertani AG. Genetic and pharmacological manipulation of urotensin II ameliorate the metabolic and atherosclerosis sequalae in mice. Arterioscler Thromb Vasc Biol 2012; 32:1809-16. [PMID: 22723440 DOI: 10.1161/atvbaha.112.252973] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Urotensin II (UII) is a potent vasoactive peptide that binds to the urotensin receptor-coupled receptor-14 (known as UT) and exerts a wide range of actions in humans and experimental animals. We tested the hypothesis that UII gene deletion or UT blockade ameliorate experimental atherosclerosis. METHODS AND RESULTS We observed a significant reduction in weight gain, visceral fat, blood pressure, circulating plasma lipids, and proatherogenic cytokines and improvement of glucose tolerance in UII knockout mice compared with wild type (P<0.05). Deletion of UII after an apolipoprotein E knockout resulted in a significant reduction in serum cytokines, adipokines, and aortic atherosclerosis compared with apolipoprotein E knockout mice. Similarly, treatment of apolipoprotein E knockout mice fed on high-fat diet with the UT antagonist SB657510A reduced weight gain, visceral fat, and hyperlipidemia and improved glucose tolerance (P<0.05) and attenuated the initiation and progression of atherosclerosis. The UT antagonist also decreased aortic extracellular signal-regulated kinase 1/2 phosphorylation and oxidant formation and serum level of cytokines (P<0.05). CONCLUSIONS These findings demonstrate for the first time the role of UII gene deletion in atherosclerosis and suggest that the use of pharmaceutical agents aimed at blocking the UII pathway may provide a novel approach in the treatment of atherosclerosis and its associated precursors such as obesity, hyperlipidemia, diabetes mellitus, and hypertension.
Collapse
Affiliation(s)
- Zhipeng You
- Division of Cardiology, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Bai Q, Zhang J, Zhang AH, Cheng LT, He L, Fan MH, Luo YJ, Wang T. Roles of human urotensin II in volume resistance hypertension in peritoneal dialysis patients. Ren Fail 2012; 34:713-7. [PMID: 22463743 DOI: 10.3109/0886022x.2012.672267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human urotensin II (hUII) is a newly discovered substance that can dilate small blood vessels to decrease the blood pressure (BP). Our previous studies showed that some volume-overloaded patients on peritoneal dialysis can maintain normal BP (congestive heart failure excluded), suggesting that these patients have volume resistance capacity. This study is to investigate whether hUII plays an important role in this subgroup of patients on peritoneal dialysis. In this study, 105 patients on continuous ambulatory peritoneal dialysis were enrolled. Volume load was evaluated by the overhydration (OH) value obtained by bioimpedance analysis. OH < 2.0 kg was defined as normal volume (NV), and OH ≥ 2.0 kg as high volume (HV). Systolic blood pressure (SBP) <130 mmHg was defined as normotension (NT) and ≥130 mmHg as hypertension (HT). The patients were thus divided into four subgroups: (1) normotension with normal volume (NT-NV), (2) normotension with high volume (NT-HV), (3) normal volume with hypertension (HT-NV), and (4) high volume with hypertension (HT-HV). hUII was measured using radioimmunoassay method. hUII was significantly higher in normal SBP group than that in high SBP group (p < 0.05). hUII was higher in the NT-HV group compared with that in the HT-HV group (p < 0.05). Our study suggests that hUII may be involved in the pathogenesis of the volume resistance HT in peritoneal dialysis patients.
Collapse
Affiliation(s)
- Qiong Bai
- Department of Nephrology, Peking University Third Hospital, Beijing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Allahverdian S, Pannu PS, Francis GA. Contribution of monocyte-derived macrophages and smooth muscle cells to arterial foam cell formation. Cardiovasc Res 2012; 95:165-72. [PMID: 22345306 DOI: 10.1093/cvr/cvs094] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Smooth muscle cells (SMCs) are the main cell type in intimal thickenings and some stages of human atherosclerosis. Like monocyte-derived macrophages, SMCs accumulate excess lipids and contribute to the total intimal foam cell population. In contrast, apolipoprotein (Apo)E-deficient and LDL receptor-deficient mice develop atherosclerotic lesions that are macrophage- as opposed to SMC-rich. The lesser contribution of SMCs to lesion development in these mouse models has distracted attention away from the importance of SMC cholesterol homeostasis in the artery wall. Intimal SMCs accumulate excess amounts of cholesteryl esters when compared with medial layer SMCs, possibly explained by reduced ATP-binding cassette transporter A1 expression and ApoA-I binding to intimal-type SMCs. The aim of this review is to compare the relative contribution of monocyte-derived macrophages and SMCs to human vs. mouse atherosclerosis, and describe what is known about lipid uptake and removal mechanisms contributing to arterial macrophage and SMC foam cell formation. An increased understanding of the contribution of these cell types to lesion development will help to delineate their relative importance in atherogenesis and as potential therapeutic targets.
Collapse
Affiliation(s)
- Sima Allahverdian
- Department of Medicine, UBC James Hogg Research Centre, Providence Heart + Lung Institute at St Paul's Hospital, Room 166, Burrard Building, 1081 Burrard Street, Vancouver, BC, Canada V6Z 1Y6
| | | | | |
Collapse
|
47
|
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: 1.3] [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
|
48
|
Nagashima M, Watanabe T, Terasaki M, Tomoyasu M, Nohtomi K, Kim-Kaneyama J, Miyazaki A, Hirano T. Native incretins prevent the development of atherosclerotic lesions in apolipoprotein E knockout mice. Diabetologia 2011; 54:2649-59. [PMID: 21786155 PMCID: PMC3168747 DOI: 10.1007/s00125-011-2241-2] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 05/31/2011] [Indexed: 01/15/2023]
Abstract
AIMS/HYPOTHESIS Several lines of evidence suggest that incretin-based therapies suppress the development of cardiovascular disease in type 2 diabetes. We investigated the possibility that glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) can prevent the development of atherosclerosis in Apoe (-/-) mice. METHODS Apoe (-/-) mice (17 weeks old) were administered GLP-1(7-36)amide, GLP-1(9-36)amide, GIP(1-42) or GIP(3-42) for 4 weeks. Aortic atherosclerosis, oxidised LDL-induced foam cell formation and related gene expression in exudate peritoneal macrophages were determined. RESULTS Administration of GLP-1(7-36)amide or GIP(1-42) significantly suppressed atherosclerotic lesions and macrophage infiltration in the aortic wall, compared with vehicle controls. These effects were cancelled by co-infusion with specific antagonists for GLP-1 and GIP receptors, namely exendin(9-39) or Pro(3)(GIP). The anti-atherosclerotic effects of GLP-1(7-36)amide and GIP(1-42) were associated with significant decreases in foam cell formation and downregulation of CD36 and acyl-coenzyme A:cholesterol acyltransferase-1 (ACAT-1) in macrophages. GLP-1 and GIP receptors were both detected in Apoe (-/-) mouse macrophages. Ex vivo incubation of macrophages with GLP-1(7-36)amide or GIP(1-42) for 48 h significantly suppressed foam cell formation. This effect was wholly abolished in macrophages pretreated with exendin(9-39) or (Pro(3))GIP, or with an adenylate cyclase inhibitor, MDL12,330A, and was mimicked by incubation with an adenylate cyclase activator, forskolin. The inactive forms, GLP-1(9-36)amide and GIP(3-42), had no effects on atherosclerosis and macrophage foam cell formation. CONCLUSIONS/INTERPRETATION Our study is the first to demonstrate that active forms of GLP-1 and GIP exert anti-atherogenic effects by suppressing macrophage foam cell formation via their own receptors, followed by cAMP activation. Molecular mechanisms underlying these effects are associated with the downregulation of CD36 and ACAT-1 by incretins.
Collapse
Affiliation(s)
- M. Nagashima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666 Japan
| | - T. Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - M. Terasaki
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666 Japan
| | - M. Tomoyasu
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666 Japan
| | - K. Nohtomi
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666 Japan
| | - J. Kim-Kaneyama
- Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan
| | - A. Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan
| | - T. Hirano
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666 Japan
| |
Collapse
|
49
|
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: 2.0] [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
|
50
|
Kiss RS, You Z, Genest J, Behm DJ, Giaid A. Urotensin II differentially regulates macrophage and hepatic cholesterol homeostasis. Peptides 2011; 32:956-63. [PMID: 21376094 DOI: 10.1016/j.peptides.2011.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 02/21/2011] [Accepted: 02/22/2011] [Indexed: 12/12/2022]
Abstract
Urotensin II (UII) is a vasoactive peptide with pleiotropic activity. Interestingly, UII levels are elevated in hyperlipidemic patients, and UII induces lipase activity in some species. However, the exact role UII plays in cholesterol homeostasis remains to be elucidated. UII knockout (UII KO) mice were generated and a plasma lipoprotein profile, and hepatocytes and macrophages cholesterol uptake, storage and synthesis was determined. UII KO had a decreased LDL cholesterol profile and liver steatosis compared to wildtype mice (WT). UII KO macrophages demonstrated enhanced ACAT activity and LDL uptake in the short term (up to 4h), of which more LDL-delivered exogenously derived cholesterol was incorporated into cholesteryl ester (CE) than the WT macrophages. UII KO macrophages generated more than two times the amount of de novo endogenously synthesized cholesterol, and of this cholesterol more than two times the relative amount was esterified to CE. In comparison, results in hepatocytes demonstrated that far more exogenously derived cholesterol was incorporated into CE in the WT cells, generating almost ten times the amount of CE than UII KO. WT cells synthesize de novo almost ten times the amount of cholesterol than UIIKO, and of that cholesterol, almost two times the amount of CE in WT than UII KO hepatocytes. In addition, more ApoB lipoproteins were secreted from WT than UII KO hepatocytes. These results demonstrate a fundamental difference between macrophages and hepatocytes in terms of cholesterol homeostasis, and suggest an important role for UII in modulating cholesterol regulation.
Collapse
Affiliation(s)
- Robert S Kiss
- Division of Cardiology, McGill University Health Center, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|