1
|
Feehan KT, Bridgewater HE, Stenkiewicz-Witeska J, De Maeyer RPH, Ferguson J, Mack M, Brown J, Ercoli G, Mawer CM, Akbar AN, Glanville JRW, Jalali P, Bracken OV, Nicolaou A, Kendall AC, Sugimoto MA, Gilroy DW. Post-resolution macrophages shape long-term tissue immunity and integrity in a mouse model of pneumococcal pneumonia. Nat Commun 2024; 15:4326. [PMID: 38773113 PMCID: PMC11109210 DOI: 10.1038/s41467-024-48138-y] [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: 10/13/2022] [Accepted: 04/19/2024] [Indexed: 05/23/2024] Open
Abstract
Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.
Collapse
Affiliation(s)
- Karen T Feehan
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Hannah E Bridgewater
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
- Centre for Sports, Exercise and Life Science, Coventry University, Priory St, Coventry, CV1 5FB, UK
| | - Jan Stenkiewicz-Witeska
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Roel P H De Maeyer
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, Windmill Road, University of Oxford, OX3 7LD, Oxford, UK
| | - John Ferguson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Matthias Mack
- Universitätsklinikum Regensburg, Innere Medizin II/Nephrologie-Transplantation, Regensburg, Germany
| | - Jeremy Brown
- UCL Respiratory, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Giuseppe Ercoli
- UCL Respiratory, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Connar M Mawer
- UCL Respiratory, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Arne N Akbar
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - James R W Glanville
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Parinaaz Jalali
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Olivia V Bracken
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Alexandra C Kendall
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Michelle A Sugimoto
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK
| | - Derek W Gilroy
- Department for Experimental and Translational Medicine, Division of Medicine, 5 University Street, University College London, London, WC1E 6JJ, UK.
| |
Collapse
|
2
|
Inazumi T, Sugimoto Y. Metabolic Regulation in Adipocytes by Prostanoid Receptors. Biol Pharm Bull 2022; 45:992-997. [DOI: 10.1248/bpb.b22-00270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tomoaki Inazumi
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yukihiko Sugimoto
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
3
|
Evans BR, Yerly A, van der Vorst EPC, Baumgartner I, Bernhard SM, Schindewolf M, Döring Y. Inflammatory Mediators in Atherosclerotic Vascular Remodeling. Front Cardiovasc Med 2022; 9:868934. [PMID: 35600479 PMCID: PMC9114307 DOI: 10.3389/fcvm.2022.868934] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/11/2022] [Indexed: 12/23/2022] Open
Abstract
Atherosclerotic vascular disease remains the most common cause of ischemia, myocardial infarction, and stroke. Vascular function is determined by structural and functional properties of the arterial vessel wall, which consists of three layers, namely the adventitia, media, and intima. Key cells in shaping the vascular wall architecture and warranting proper vessel function are vascular smooth muscle cells in the arterial media and endothelial cells lining the intima. Pathological alterations of this vessel wall architecture called vascular remodeling can lead to insufficient vascular function and subsequent ischemia and organ damage. One major pathomechanism driving this detrimental vascular remodeling is atherosclerosis, which is initiated by endothelial dysfunction allowing the accumulation of intimal lipids and leukocytes. Inflammatory mediators such as cytokines, chemokines, and modified lipids further drive vascular remodeling ultimately leading to thrombus formation and/or vessel occlusion which can cause major cardiovascular events. Although it is clear that vascular wall remodeling is an elementary mechanism of atherosclerotic vascular disease, the diverse underlying pathomechanisms and its consequences are still insufficiently understood.
Collapse
Affiliation(s)
- Bryce R. Evans
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Anaïs Yerly
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Iris Baumgartner
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sarah Maike Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marc Schindewolf
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- *Correspondence: Yvonne Döring
| |
Collapse
|
4
|
Tanaka N, Yamaguchi H, Mano N. Involvement of H +-gradient dependent transporter in PGE 2 release from A549 cells. Prostaglandins Leukot Essent Fatty Acids 2019; 149:30-36. [PMID: 31421525 DOI: 10.1016/j.plefa.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/18/2019] [Accepted: 08/07/2019] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to identify the transporter involved in the release of prostaglandin E2 (PGE2). In the present study, transport assays were conducted using membrane vesicles prepared from human lung adenocarcinoma A549 cells, thus enabling identification of the novel exporter present in A549 cells. PGE2 transport into A549 vesicles was higher in the presence of a proton (H+)-gradient, thus suggesting the involvement of PGE2H+ symporter in PGE2 transport. Results from our experiments showed enhanced PGE2 release in A549 cells in the presence of H+-gradient ([H+]extracellular < [H+]intracellular). Moreover, in vesicular transport assays, H+-gradient-dependent transport of PGE2 did not show saturation up to 500 μM PGE2, and 10 mM aromatic monocarboxylic acids (acetylsalicylic acid, salicylic acid, and p-nitrobenzoic acid) significantly inhibited PGE2 transport by 62-70%. These results suggest, the involvement of monocarboxylate transporters in the H+-gradient-dependent PGE2 export.
Collapse
Affiliation(s)
- Nobuaki Tanaka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Hiroaki Yamaguchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, 980-8574, Japa; Yamagata University Graduate School of Medicine/Department of Pharmacy, Yamagata University Hospital, 2-2-2, Iida-nishi, Yamagata, 990-9585, Japan.
| | - Nariyasu Mano
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, 980-8574, Japa
| |
Collapse
|
5
|
Hardy E, Hardy-Sosa A, Fernandez-Patron C. MMP-2: is too low as bad as too high in the cardiovascular system? Am J Physiol Heart Circ Physiol 2018; 315:H1332-H1340. [DOI: 10.1152/ajpheart.00198.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Matrix metalloproteinase (MMP)-2 cleaves a broad spectrum of substrates, including extracellular matrix components (responsible for normal tissue remodeling) and cytokines (modulators of the inflammatory response to physiological insults such as tissue damage). MMP-2 expression is elevated in many cardiovascular pathologies (e.g., myocardial infarction, hypertensive heart disease) where tissue remodeling and inflammatory responses are perturbed. Thus, it has generally been assumed that blockade of MMP-2 activity will yield therapeutic effects. Here, we provide a counterargument to this dogma based on 1) preclinical studies on Mmp2-null ( Mmp2−/−) mice and 2) clinical studies on patients with inactivating MMP2 gene mutations. Furthermore, we put forward the hypothesis that, when MMP-2 activity falls below baseline, the bioavailability of proinflammatory cytokines normally cleaved and inactivated by MMP-2 increases, leading to the production of cytokines and cardiac secretion of phospholipase A2activity into the circulation, which stimulate systemic inflammation that perturbs lipid metabolism in target organs. Finally, we suggest that insufficient understanding of the consequences of MMP-2 deficiency remains a major factor in the failure of MMP-2 inhibitor-based therapeutic approaches. This paucity of knowledge precludes our ability to effectively intervene in cardiovascular and noncardiovascular pathologies at the level of MMP-2.
Collapse
Affiliation(s)
- Eugenio Hardy
- Biotechnology Laboratory, Study Center for Research and Biological Evaluations, Institute of Pharmacy and Foods, University of Havana, Havana, Cuba
| | | | - Carlos Fernandez-Patron
- Department of Biochemistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Avendaño MS, García-Redondo AB, Zalba G, González-Amor M, Aguado A, Martínez-Revelles S, Beltrán LM, Camacho M, Cachofeiro V, Alonso MJ, Salaices M, Briones AM. mPGES-1 (Microsomal Prostaglandin E Synthase-1) Mediates Vascular Dysfunction in Hypertension Through Oxidative Stress. Hypertension 2018; 72:492-502. [PMID: 29891646 DOI: 10.1161/hypertensionaha.118.10833] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/28/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022]
Abstract
mPGES-1 (microsomal prostaglandin E synthase-1), the downstream enzyme responsible for PGE2 (prostaglandin E2) synthesis in inflammatory conditions and oxidative stress are increased in vessels from hypertensive animals. We evaluated the role of mPGES-1-derived PGE2 in the vascular dysfunction and remodeling in hypertension and the possible contribution of oxidative stress. We used human peripheral blood mononuclear cells from asymptomatic patients, arteries from untreated and Ang II (angiotensin II)-infused mPGES-1-/- and mPGES-1+/+ mice, and vascular smooth muscle cells exposed to PGE2 In human cells, we found a positive correlation between mPGES-1 mRNA and carotid intima-media thickness (r=0.637; P<0.001) and with NADPH oxidase-dependent superoxide production (r=0.417; P<0.001). In Ang II-infused mice, mPGES-1 deletion prevented all of the following: (1) the augmented wall:lumen ratio, vascular stiffness, and altered elastin structure; (2) the increased gene expression of profibrotic and proinflammatory markers; (3) the increased vasoconstrictor responses and endothelial dysfunction; (4) the increased NADPH oxidase activity and the diminished mitochondrial membrane potential; and (5) the increased reactive oxygen species generation and reduced NO bioavailability. In vascular smooth muscle cells or aortic segments, PGE2 increased NADPH oxidase expression and activity and reduced mitochondrial membrane potential, effects that were abolished by antagonists of the PGE2 receptors (EP), EP1 and EP3, and by JNK (c-Jun N-terminal kinase) and ERK1/2 (extracellular-signal-regulated kinases 1/2) inhibition. Deletion of mPGES-1 augmented vascular production of PGI2 suggesting rediversion of the accumulated PGH2 substrate. In conclusion, mPGES-1-derived PGE2 is involved in vascular remodeling, stiffness, and endothelial dysfunction in hypertension likely through an increase of oxidative stress produced by NADPH oxidase and mitochondria.
Collapse
Affiliation(s)
- María S Avendaño
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.)
| | - Ana B García-Redondo
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.).,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| | - Guillermo Zalba
- Departamento de Bioquímica y Genética, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, Pamplona, Spain (G.Z.)
| | - María González-Amor
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.)
| | - Andrea Aguado
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.)
| | - Sonia Martínez-Revelles
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.).,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| | - Luis M Beltrán
- Unidad Clínico-Experimental de Riesgo Vascular-Medicina Interna, Instituto de Biomedicina de Sevilla, Hospital Virgen del Rocío, Spain (L.M.B.)
| | - Mercedes Camacho
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.).,Laboratorio de Angiología, Biología Vascular e Inflamación, Instituto de Investigación Biomédica Sant Pau, Barcelona, Spain (M.C.)
| | - Victoria Cachofeiro
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.).,Departamento de Fisiología, Facultad de Medicina, Instituto de Investigación Gregorio Marañón, Universidad Complutense de Madrid, Spain (V.C.)
| | - María J Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.).,Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain (M.J.A.)
| | - Mercedes Salaices
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.).,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| | - Ana M Briones
- From the Departmento de Farmacología, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, Spain (M.S.A., A.B.G.-R., M.G.-A., A.A., S.M.-R., M.S., A.M.B.) .,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain (A.B.G.-R., S.M.-R., M.C., V.C., M.J.A., M.S., A.M.B.)
| |
Collapse
|
7
|
Abstract
As the largest receptor gene family in the human genome, with >800 members, the signal-transducing G protein-coupled receptors (GPCRs) play critical roles in nearly all conceivable physiological processes, ranging from the sensing of photons and odorants to metabolic homeostasis and migration of leukocytes. Unfortunately, an exhaustive review of the several hundred GPCRs expressed by myeloid cells/macrophages (P.J. Groot-Kormelink, L .Fawcett, P.D. Wright, M. Gosling, and T.C. Kent, BMC Immunol 12:57, 2012, doi:10.1186/1471-2172-13-57) is beyond the scope of this chapter; however, we will endeavor to cover the GPCRs that contribute to the major facets of macrophage biology, i.e., those whose expression is restricted to macrophages and the GPCRs involved in macrophage differentiation/polarization, microbial elimination, inflammation and resolution, and macrophage-mediated pathology. The chemokine receptors, a major group of myeloid GPCRs, will not be extensively covered as they are comprehensively reviewed elsewhere.
Collapse
|
8
|
Decreased vasorelaxation induced by iloprost during acute inflammation in human internal mammary artery. Eur J Pharmacol 2017; 804:31-37. [PMID: 28373136 DOI: 10.1016/j.ejphar.2017.03.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 12/25/2022]
Abstract
Cyclooxygenase-2 (COX-2) induction in human internal mammary arteries (IMA) under inflammatory conditions has been associated with attenuated norepinephrine (NE)-induced vasoconstriction. This effect was associated with increased prostaglandin (PG) E2 and prostacyclin (PGI2) releases. The present study was designed to assess the role of these PG and their receptors (EP and IP, respectively) on the vascular reactivity during acute inflammation. Isolated IMA were cultured in the absence (Control conditions) or presence (Inflammatory conditions) of both interleukin-1 beta (IL-1β) and lipopolysaccharide (LPS). The vasorelaxation and the increased content of cyclic adenosine monophosphate (cAMP) induced by iloprost, a PGI2 analogue, were significantly reduced under inflammatory conditions and restored in preparations cultured with the IP antagonist (CAY10441). Decreased cAMP levels under inflammatory conditions are due to at least increased phosphodiesterase (PDE) 4B expression. On the other hand, PGE2, thromboxane analogues and EP agonists-induced vasoconstrictions were not affected under inflammatory conditions. No vasorelaxation was observed with PGD2, PGE2 or the EP2/4 agonists in pre-contracted IMA. Finally, using RT-qPCR and immunohistochemistry, the COX-2, IP receptor and PGI2 synthase (PGIS) were detected. A significant increase of COX-2 and moderate increase of IP mRNA expression was observed under inflammatory conditions, whereas PGIS mRNA level was not affected. This study demonstrates that PGI2/IP receptor signalling and PGI2-induced relaxation are impaired in human IMA during acute inflammation, whereas the responses induced by other prostanoids are not affected. These results could explain some of the mechanisms of vascular dysfunction reported in inflammatory conditions.
Collapse
|
9
|
Prostanoids in the pathophysiology of human coronary artery. Prostaglandins Other Lipid Mediat 2017; 133:20-28. [PMID: 28347710 DOI: 10.1016/j.prostaglandins.2017.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/16/2017] [Accepted: 03/23/2017] [Indexed: 01/16/2023]
Abstract
Coronary artery disease is one of the leading causes of death in wordwide. There is growing evidence that prostanoids are involved in the physiology and pathophysiology of the human coronary artery by controlling vascular tone, remodelling of the vascular wall or angiogenesis. In this review, the production of prostanoids and the expression of prostanoid receptors in human coronary artery in health or disease are described. In addition, the interactions between sex hormones and prostanoids, their participations in the development of coronary artery diseases have been addressed. Globally, most of the studies performed in human coronary artery preparations have shown that prostacyclin (PGI2) has beneficial effects by inducing vasodilatation and promoting angiogenesis while reverse effects are confirmed by thromboxane A2 (TxA2). More studies are needed to determine the roles of the other prostanoids (PGE2, PGD2 and PGF2α) in vascular functions of the human coronary artery. Finally, in addition to the in vitro data about the human coronary artery, myocardial infarction induced by cyclooxygenase-2 (COX-2) inhibitor and the protective effects of aspirin after coronary artery bypass surgery suggest that prostanoids are key mediators in coronary homeostasis.
Collapse
|
10
|
Avendaño MS, Martínez-Revelles S, Aguado A, Simões MR, González-Amor M, Palacios R, Guillem-Llobat P, Vassallo DV, Vila L, García-Puig J, Beltrán LM, Alonso MJ, Cachofeiro MV, Salaices M, Briones AM. Role of COX-2-derived PGE2 on vascular stiffness and function in hypertension. Br J Pharmacol 2016; 173:1541-55. [PMID: 26856544 DOI: 10.1111/bph.13457] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 01/08/2016] [Accepted: 01/29/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Prostanoids derived from COX-2 and EP receptors are involved in vascular remodelling in different cardiovascular pathologies. This study evaluates the contribution of COX-2 and EP1 receptors to vascular remodelling and function in hypertension. EXPERIMENTAL APPROACH Spontaneously hypertensive rats (SHR) and angiotensin II (AngII)-infused (1.44 mg · kg(-1) · day(-1), 2 weeks) mice were treated with the COX-2 inhibitor celecoxib (25 mg · kg(-1) · day(-1) i.p) or with the EP1 receptor antagonist SC19220 (10 mg · kg(-1) · day(-1) i.p.). COX-2(-/-) mice with or without AngII infusion were also used. KEY RESULTS Celecoxib and SC19220 treatment did not modify the altered lumen diameter and wall : lumen ratio in mesenteric resistance arteries from SHR-infused and/or AngII-infused animals. However, both treatments and COX-2 deficiency decreased the augmented vascular stiffness in vessels from hypertensive animals. This was accompanied by diminished vascular collagen deposition, normalization of altered elastin structure and decreased connective tissue growth factor and plasminogen activator inhibitor-1 gene expression. COX-2 deficiency and SC19220 treatment diminished the increased vasoconstrictor responses and endothelial dysfunction induced by AngII infusion. Hypertensive animals showed increased mPGES-1 expression and PGE2 production in vascular tissue, normalized by celecoxib. Celecoxib treatment also decreased AngII-induced macrophage infiltration and TNF-α expression. Macrophage conditioned media (MCM) increased COX-2 and collagen type I expression in vascular smooth muscle cells; the latter was reduced by celecoxib treatment. CONCLUSIONS AND IMPLICATIONS COX-2 and EP1 receptors participate in the increased extracellular matrix deposition and vascular stiffness, the impaired vascular function and inflammation in hypertension. Targeting PGE2 receptors might have benefits in hypertension-associated vascular damage.
Collapse
Affiliation(s)
- M S Avendaño
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - S Martínez-Revelles
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - A Aguado
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - M R Simões
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain.,Dept. Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - M González-Amor
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - R Palacios
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - P Guillem-Llobat
- Centro de Biología Molecular "Severo Ochoa", UAM-CSIC, Madrid, Spain
| | - D V Vassallo
- Dept. Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - L Vila
- Laboratorio de Angiología, Biología Vascular e Inflamación, Instituto de Investigación Biomédica (IIB Sant Pau), Barcelona, Spain
| | - J García-Puig
- Servicio de Medicina Interna, Hospital Universitario La Paz, UAM, IdiPaz, Madrid, Spain
| | - L M Beltrán
- Servicio de Medicina Interna, Hospital Universitario La Paz, UAM, IdiPaz, Madrid, Spain
| | - M J Alonso
- Dept Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - M V Cachofeiro
- Dept. Fisiología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - M Salaices
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - A M Briones
- Dept. Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
11
|
|
12
|
Zhao Y, Vanhoutte PM, Leung SWS. α1 -Adrenoceptor activation of PKC-ε causes heterologous desensitization of thromboxane receptors in the aorta of spontaneously hypertensive rats. Br J Pharmacol 2015; 172:3687-701. [PMID: 25857252 DOI: 10.1111/bph.13157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/19/2015] [Accepted: 03/31/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE In the aorta of adult spontaneously hypertensive (SHR), but not in that of normotensive Wistar-Kyoto (WKY), rats, previous exposure to phenylephrine inhibits subsequent contractions to PGE2 . The present experiments were designed to examine the mechanism(s) underlying this inhibition. EXPERIMENTAL APPROACH Isometric tension was measured in isolated rings of SHR and WKY aortae. Gene expression and protein presence were measured by quantitative real-time PCR and Western blotting respectively. KEY RESULTS In aorta of 18 weeks SHR, but not age-matched WKY, pre-exposure to phenylephrine inhibited subsequent contractions to PGE2 that were mediated by thromboxane prostanoid (TP) receptors. This inhibition was not observed in preparations of pre-hypertensive 5-week-old SHR, and was significantly larger in those of 36- than 18-week-old SHR. Pre-exposure to the PKC activator, phorbol 12,13-dibutyrate, also inhibited subsequent contractions to PGE2 in SHR aortae. The selective inhibitor of PKC-ε, ε-V1-2, abolished the desensitization caused by pre-exposure to phenylephrine. Two molecular PKC bands were detected and their relative intensities differed in 36-week-old WKY and SHR vascular smooth muscle. The mRNA expressions of PKC-α, PKC-ε, PK-N2 and PKC-ζ and of G protein-coupled kinase (GRK)-2, GRK4 and β-arrestin2 were higher in SHR than WKY aortae. CONCLUSIONS AND IMPLICATIONS These experiments suggest that in the SHR but not the WKY aorta, α1 -adrenoceptor activation desensitizes TP receptors through activation of PKC-ε. This heterologous desensitization is a consequence of the chronic exposure to high arterial pressure.
Collapse
Affiliation(s)
- Yingzi Zhao
- Department of Pharmacology & Pharmacy and Stake Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Paul M Vanhoutte
- Department of Pharmacology & Pharmacy and Stake Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Susan W S Leung
- Department of Pharmacology & Pharmacy and Stake Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
13
|
Torres-Atencio I, Ainsua-Enrich E, de Mora F, Picado C, Martín M. Prostaglandin E2 prevents hyperosmolar-induced human mast cell activation through prostanoid receptors EP2 and EP4. PLoS One 2014; 9:e110870. [PMID: 25329458 PMCID: PMC4203853 DOI: 10.1371/journal.pone.0110870] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/24/2014] [Indexed: 11/26/2022] Open
Abstract
Background Mast cells play a critical role in allergic and inflammatory diseases, including exercise-induced bronchoconstriction (EIB) in asthma. The mechanism underlying EIB is probably related to increased airway fluid osmolarity that activates mast cells to the release inflammatory mediators. These mediators then act on bronchial smooth muscle to cause bronchoconstriction. In parallel, protective substances such as prostaglandin E2 (PGE2) are probably also released and could explain the refractory period observed in patients with EIB. Objective This study aimed to evaluate the protective effect of PGE2 on osmotically activated mast cells, as a model of exercise-induced bronchoconstriction. Methods We used LAD2, HMC-1, CD34-positive, and human lung mast cell lines. Cells underwent a mannitol challenge, and the effects of PGE2 and prostanoid receptor (EP) antagonists for EP1–4 were assayed on the activated mast cells. Beta-hexosaminidase release, protein phosphorylation, and calcium mobilization were assessed. Results Mannitol both induced mast cell degranulation and activated phosphatidyl inositide 3-kinase and mitogen-activated protein kinase (MAPK) pathways, thereby causing de novo eicosanoid and cytokine synthesis. The addition of PGE2 significantly reduced mannitol-induced degranulation through EP2 and EP4 receptors, as measured by beta-hexosaminidase release, and consequently calcium influx. Extracellular-signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38 phosphorylation were diminished when compared with mannitol activation alone. Conclusions Our data show a protective role for the PGE2 receptors EP2 and EP4 following osmotic changes, through the reduction of human mast cell activity caused by calcium influx impairment and MAP kinase inhibition.
Collapse
MESH Headings
- Asthma, Exercise-Induced/genetics
- Asthma, Exercise-Induced/metabolism
- Asthma, Exercise-Induced/pathology
- Cell Degranulation
- Cell Line
- Dinoprostone/genetics
- Dinoprostone/metabolism
- Diuretics, Osmotic/pharmacology
- Extracellular Signal-Regulated MAP Kinases/genetics
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Humans
- Lung/metabolism
- Lung/pathology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/genetics
- Mannitol/pharmacology
- Mast Cells/metabolism
- Mast Cells/pathology
- Osmotic Pressure
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
Collapse
Affiliation(s)
- Ivonne Torres-Atencio
- Unidad de Farmacología, Facultad de Medicina, Universidad de Panamá, Panama, Panama Republic
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Erola Ainsua-Enrich
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Unitat de Bioquímica i Biologia Molecular, Department de Ciències Fisològiques I, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Fernando de Mora
- Department de Farmacologia, Terapéutica i Toxicologia, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - César Picado
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Margarita Martín
- Laboratori d'Immunoal·lèrgia Respiratòria Clínica i Experimental, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Unitat de Bioquímica i Biologia Molecular, Department de Ciències Fisològiques I, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
14
|
García-Alonso V, Clària J. Prostaglandin E2 signals white-to-brown adipogenic differentiation. Adipocyte 2014; 3:290-6. [PMID: 26317053 DOI: 10.4161/adip.29993] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 02/07/2023] Open
Abstract
The formation of new adipocytes from precursor cells is a crucial aspect of normal adipose tissue function. During the adipogenic process, adipocytes differentiated from mesenchymal stem cells give rise to two main types of fat: white adipose tissue (WAT) characterized by the presence of adipocytes containing large unilocular lipid droplets, and brown adipose tissue (BAT) composed by multilocular brown adipocytes packed with mitochondria. WAT is not only important for energy storage but also as an endocrine organ regulating whole body homeostasis by secreting adipokines and other mediators, which directly impact metabolic functions in obesity. By contrast, BAT is specialized in dissipating energy in form of heat and has salutary effects in combating obesity and associated disorders. Unfortunately, WAT is the predominant fat type, whereas BAT is scarce and located in discrete pockets in adult humans. Luckily, another type of brown adipocytes, called beige or brite (brown-in-white) adipocytes, with similar functions to those of "classical" brown adipocytes has recently been identified in WAT. In this review, a close look is given into the role of bioactive lipid mediators in the regulation of adipogenesis, with a special emphasis on the role of the microsomal prostaglandin E (PGE) synthase-1, a terminal enzyme in PGE2 biosynthesis, as a key regulator of white-to-brown adipogenesis in WAT.
Collapse
|
15
|
Gomez I, Benyahia C, Louedec L, Leséche G, Jacob MP, Longrois D, Norel X. Decreased PGE₂ content reduces MMP-1 activity and consequently increases collagen density in human varicose vein. PLoS One 2014; 9:e88021. [PMID: 24505358 PMCID: PMC3914898 DOI: 10.1371/journal.pone.0088021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 01/02/2014] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED Varicose veins are elongated and dilated saphenous veins. Despite the high prevalence of this disease, its pathogenesis remains unclear. AIMS In this study, we investigated the control of matrix metalloproteinases (MMPs) expression by prostaglandin (PG)E₂ during the vascular wall remodeling of human varicose veins. METHODS AND RESULTS Varicose (small (SDv) and large diameter (LDv)) and healthy saphenous veins (SV) were obtained after surgery. Microsomal and cytosolic PGE-synthases (mPGES and cPGES) protein and mRNA responsible for PGE₂ metabolism were analyzed in all veins. cPGES protein was absent while its mRNA was weakly expressed. mPGES-2 expression was similar in the different saphenous veins. mPGES-1 mRNA and protein were detected in healthy veins and a significant decrease was found in LDv. Additionally, 15-hydroxyprostaglandin dehydrogenase (15-PGDH), responsible for PGE₂ degradation, was over-expressed in varicose veins. These variations in mPGES-1 and 15-PGDH density account for the decreased PGE₂ level observed in varicose veins. Furthermore, a significant decrease in PGE₂ receptor (EP4) levels was also found in SDv and LDv. Active MMP-1 and total MMP-2 concentrations were significantly decreased in varicose veins while the tissue inhibitors of metalloproteinases (TIMP -1 and -2), were significantly increased, probably explaining the increased collagen content found in LDv. Finally, the MMP/TIMP ratio is restored by exogenous PGE₂ in varicose veins and reduced in presence of an EP4 receptor antagonist in healthy veins. CONCLUSIONS In conclusion, PGE₂ could be responsible for the vascular wall thickening in human varicose veins. This mechanism could be protective, strengthening the vascular wall in order to counteract venous stasis.
Collapse
Affiliation(s)
- Ingrid Gomez
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
| | - Chabha Benyahia
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
| | | | - Guy Leséche
- INSERM, U698, Paris, France
- AP-HP CHU X. Bichat, Department of Vascular and Thoracic Surgery, University Paris Diderot, Sorbonne Paris-Cité, UMR-S698, Paris, France
| | | | - Dan Longrois
- INSERM, U698, Paris, France
- AP-HP CHU X. Bichat, Department of Anesthesia and Intensive Care, University Paris Diderot, Sorbonne Paris-Cité, UMR-S698, Paris, France
| | - Xavier Norel
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
- * E-mail:
| |
Collapse
|
16
|
De Caterina R. Inhibiting thrombosis without causing bleeding: can EP3 blockers fulfil the dream? Cardiovasc Res 2014; 101:335-8. [DOI: 10.1093/cvr/cvu020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
17
|
Cutler C, Multani P, Robbins D, Kim HT, Le T, Hoggatt J, Pelus LM, Desponts C, Chen YB, Rezner B, Armand P, Koreth J, Glotzbecker B, Ho VT, Alyea E, Isom M, Kao G, Armant M, Silberstein L, Hu P, Soiffer RJ, Scadden DT, Ritz J, Goessling W, North TE, Mendlein J, Ballen K, Zon LI, Antin JH, Shoemaker DD. Prostaglandin-modulated umbilical cord blood hematopoietic stem cell transplantation. Blood 2013; 122:3074-81. [PMID: 23996087 PMCID: PMC3811179 DOI: 10.1182/blood-2013-05-503177] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/20/2013] [Indexed: 01/16/2023] Open
Abstract
Umbilical cord blood (UCB) is a valuable source of hematopoietic stem cells (HSCs) for use in allogeneic transplantation. Key advantages of UCB are rapid availability and less stringent requirements for HLA matching. However, UCB contains an inherently limited HSC count, which is associated with delayed time to engraftment, high graft failure rates, and early mortality. 16,16-Dimethyl prostaglandin E2 (dmPGE2) was previously identified to be a critical regulator of HSC homeostasis, and we hypothesized that brief ex vivo modulation with dmPGE2 could improve patient outcomes by increasing the "effective dose" of HSCs. Molecular profiling approaches were used to determine the optimal ex vivo modulation conditions (temperature, time, concentration, and media) for use in the clinical setting. A phase 1 trial was performed to evaluate the safety and therapeutic potential of ex vivo modulation of a single UCB unit using dmPGE2 before reduced-intensity, double UCB transplantation. Results from this study demonstrated clear safety with durable, multilineage engraftment of dmPGE2-treated UCB units. We observed encouraging trends in efficacy, with accelerated neutrophil recovery (17.5 vs 21 days, P = .045), coupled with preferential, long-term engraftment of the dmPGE2-treated UCB unit in 10 of 12 treated participants.
Collapse
|
18
|
Gomez I, Foudi N, Longrois D, Norel X. The role of prostaglandin E2 in human vascular inflammation. Prostaglandins Leukot Essent Fatty Acids 2013; 89:55-63. [PMID: 23756023 DOI: 10.1016/j.plefa.2013.04.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 10/26/2022]
Abstract
Prostaglandins (PG) are the product of a cascade of enzymes such as cyclooxygenases and PG synthases. Among PG, PGE2 is produced by 3 isoforms of PGE synthase (PGES) and through activation of its cognate receptors (EP1-4), this PG is involved in the pathophysiology of vascular diseases. Some anti-inflammatory drugs (e.g. glucocorticoids, nonsteroidal anti-inflammatory drugs) interfere with its metabolism or effects. Vascular cells can initiate many of the responses associated with inflammation. In human vascular tissue, PGE2 is involved in many physiological processes, such as increasing vascular permeability, cell proliferation, cell migration and control of vascular smooth muscle tone. PGE2 has been shown to contribute to the pathogenesis of atherosclerosis, abdominal aortic aneurysm but also in physiologic/adaptive processes such as angiogenesis. Understanding the roles of PGE2 and its cognate receptors in vascular diseases could help to identify diagnostic and prognostic biomarkers. In addition, from these recent studies new promising therapeutic approaches like mPGES-1 inhibition and/or EP4-antagonism should be investigated.
Collapse
Affiliation(s)
- I Gomez
- INSERM, U698, Paris F-75018, France; University Paris Nord, UMR-S698, Paris F-75018, France
| | | | | | | |
Collapse
|
19
|
Affiliation(s)
- Francesco Cipollone
- From the European Center of Excellence on Atherosclerosis, Hypertension and Dyslipidemia, and Clinical Research Center, Center of Excellence on Aging (Ce.S.I.), G. d’Annunzio University, Chieti-Pescara, Italy
| | - Donato Santovito
- From the European Center of Excellence on Atherosclerosis, Hypertension and Dyslipidemia, and Clinical Research Center, Center of Excellence on Aging (Ce.S.I.), G. d’Annunzio University, Chieti-Pescara, Italy
| |
Collapse
|
20
|
Ozen G, Topal G, Gomez I, Ghorreshi A, Boukais K, Benyahia C, Kanyinda L, Longrois D, Teskin O, Uydes-Dogan BS, Norel X. Control of human vascular tone by prostanoids derived from perivascular adipose tissue. Prostaglandins Other Lipid Mediat 2013; 107:13-7. [PMID: 23791663 DOI: 10.1016/j.prostaglandins.2013.06.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 05/23/2013] [Accepted: 06/07/2013] [Indexed: 11/16/2022]
Abstract
Perivascular adipose tissue (PVAT) surrounds most vessels and has now been recognized as a regulator of vascular functions. This effect of PVAT has been mostly demonstrated in vessels obtained from rats and mice. Thus, the aim of this study was to investigate anti-contractile effect of PVAT surrounding human coronary bypass grafts such as saphenous vein (SV) and internal mammary artery (IMA). Moreover, we aimed to determine the involvement of prostanoids in the anticontractile effect of PVAT. Human SV and IMA preparations were set up in an organ bath. The presence of PVAT in SV and IMA preparations significantly attenuated the contractile response to noradrenaline (NA). Preincubation with indomethacin, a cyclooxygenase inhibitor, increased NA contraction in SV preparations with PVAT. This effect was not observed in IMA preparation with PVAT incubated with indomethacin. The lower measurements of prostaglandin E2 (PGE2) released from PVAT surrounding IMA versus SV supported these effects. In conclusion, our results show that PVAT of SV could attenuate NA-induced contraction by releasing both PGE2 and prostacyclin (PGI2). In contrast to SV, PVAT of IMA exerts its anti-contractile effect independently from prostanoids. These observations suggest that retaining PVAT in human SV and IMA preparations may have potential clinical implications to improve coronary bypass graft patency.
Collapse
Affiliation(s)
- Gulsev Ozen
- Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|