1
|
Prasongtanakij S, Soontrapa K, Thumkeo D. The role of prostanoids in regulatory T cells and their implications in inflammatory diseases and cancers. Eur J Cell Biol 2025; 104:151482. [PMID: 40184828 DOI: 10.1016/j.ejcb.2025.151482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
Regulatory T cells (Tregs) play an important role in the immune system through the regulation of immunological self-tolerance and homeostasis. Furthermore, increasing evidence suggests the potential contribution of Tregs beyond immunity in the process of repairing various injured tissues. Tregs are generally characterised by the constitutive expression of forkhead box protein 3 (FOXP3) transcription factor in the nucleus and high expression levels of CD25 and CTLA-4 on the cell surface. To date, a large number of molecules have been identified as key regulators of Treg differentiation and function. Among these molecules are prostanoids, which are multifaceted lipid mediators. Prostanoids are produced from arachidonic acid through the catalytic activity of the enzyme cyclooxygenase and exert their functions through the 9 cognate receptors, DP1-2, EP1-EP4, FP, IP and TP. We briefly review previous studies on the regulatory mechanism of Tregs and then discuss recent works on the modulatory role of prostanoids.
Collapse
Affiliation(s)
- Somsak Prasongtanakij
- Laboratory of Immunopharmacology, Kyoto University Graduate School of Medicine, Japan
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Dean Thumkeo
- Laboratory of Immunopharmacology, Kyoto University Graduate School of Medicine, Japan; Center for Medical Education and Internationalization, Kyoto University Faculty of Medicine, Japan.
| |
Collapse
|
2
|
Tanaka M, Szatmári I, Vécsei L. Quinoline Quest: Kynurenic Acid Strategies for Next-Generation Therapeutics via Rational Drug Design. Pharmaceuticals (Basel) 2025; 18:607. [PMID: 40430428 PMCID: PMC12114834 DOI: 10.3390/ph18050607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Quinoline-derived metabolites exhibit notable chemical complexity. What causes minor structural alterations to induce significant changes in disease outcomes? Historically, eclipsed by more straightforward scaffolds, these chemicals serve as a dynamic hub in tryptophan metabolism, linking immunomodulation, excitotoxicity, and cancer. However, many of these compounds struggle to cross the blood-brain barrier, and we still do not fully understand how certain structural changes affect their bioavailability or off-target effects. Thus, contemporary research highlights halogenation, esterification, and computational modeling to enhance structure-activity relationships. SUMMARY This narrative review emphasizes the integration of rational drug design, multi-target ligands, and prodrug methods in enhancing quinoline scaffolds. We explore each molecule's therapeutic promise, refine each scaffold's design, and develop each derivative to maximize clinical utility. Translating these laboratory findings into clinical practice, however, remains a formidable challenge. CONCLUSIONS Through the synthesis of findings regarding NMDA receptor antagonism, improved oral bioavailability, and reduced metabolic instability, we demonstrate how single-site changes might modulate excitotoxicity and immunological signaling. Advancing quinoline-based medicines will yield significant advancements in neurology, psychiatry, and oncology. This enlarged framework fosters collaborative discovery, engages various audiences, and advances the field towards next-generation disease-modifying therapies. Robust preclinical validation, patient classification, and comprehensive toxicity evaluations are crucial stages for achieving these extensive endeavors and fostering future therapeutic discoveries globally.
Collapse
Affiliation(s)
- Masaru Tanaka
- Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - István Szatmári
- Institute of Pharmaceutical Chemistry and HUN-REN–SZTE Stereochemistry Research Group, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
| | - László Vécsei
- Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
3
|
Javaid A, Mudavath SL. Niacin-induced flushing: Mechanism, pathophysiology, and future perspectives. Arch Biochem Biophys 2024; 761:110163. [PMID: 39322100 DOI: 10.1016/j.abb.2024.110163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/06/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Flushing is a typical physiological reaction to high emotional reactions. It is characterized by cutaneous vasodilation and a feeling of warmth and skin redness, especially in the face areas. Flushing is frequently linked to social anxiety, but it can also be a sign of a number of benign and malignant medical disorders. The study focuses on niacin-induced flushing, a well-researched side effect of the niacin, a drug which increases cholesterol levels. Niacin-induced flushing occurs when the hydroxycarboxylic acid receptor 2 (HCA2 or GPR109A) is activated. This starts a signaling cascade that releases prostaglandins, especially PGD2, which causes cutaneous vasodilation. Furthermore, niacin directly interacts with the transient receptor potential (TRP) channel TRPV1, offering a different, non-prostaglandin-based explanation for flushing brought on by niacin, highlighting the intricate physiological mechanisms behind this widespread occurrence. The review delves deeper into the advantages of niacin treatment for the cardiovascular system, highlighting how it can improve lipid profiles and lower cardiovascular events when used with statins. To sum it up, this study offers a thorough understanding of flushing, including its physiological foundation, many etiologies, diagnostic difficulties, and the subtleties of flushing caused by niacin. The investigation of innovative dose forms and nanomedicine highlights the continuous endeavors to improve patient compliance and reduce side effects, laying the groundwork for further developments in flushing treatment.
Collapse
Affiliation(s)
- Aaqib Javaid
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab, 140306, India
| | - Shyam Lal Mudavath
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab, 140306, India; Department of Animal Biology, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli Hyderabad, 500046, Telangana, India.
| |
Collapse
|
4
|
Renaud D, Höller A, Michel M. Potential Drug-Nutrient Interactions of 45 Vitamins, Minerals, Trace Elements, and Associated Dietary Compounds with Acetylsalicylic Acid and Warfarin-A Review of the Literature. Nutrients 2024; 16:950. [PMID: 38612984 PMCID: PMC11013948 DOI: 10.3390/nu16070950] [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: 03/05/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
In cardiology, acetylsalicylic acid (ASA) and warfarin are among the most commonly used prophylactic therapies against thromboembolic events. Drug-drug interactions are generally well-known. Less known are the drug-nutrient interactions (DNIs), impeding drug absorption and altering micronutritional status. ASA and warfarin might influence the micronutritional status of patients through different mechanisms such as binding or modification of binding properties of ligands, absorption, transport, cellular use or concentration, or excretion. Our article reviews the drug-nutrient interactions that alter micronutritional status. Some of these mechanisms could be investigated with the aim to potentiate the drug effects. DNIs are seen occasionally in ASA and warfarin and could be managed through simple strategies such as risk stratification of DNIs on an individual patient basis; micronutritional status assessment as part of the medical history; extensive use of the drug-interaction probability scale to reference little-known interactions, and application of a personal, predictive, and preventive medical model using omics.
Collapse
Affiliation(s)
- David Renaud
- DIU MAPS, Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
- DIU MAPS, Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
- Fundacja Recover, 05-124 Skrzeszew, Poland
| | - Alexander Höller
- Department of Nutrition and Dietetics, University Hospital Innsbruck, 6020 Innsbruck, Austria
| | - Miriam Michel
- Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
5
|
Zeng C, Liu J, Zheng X, Hu X, He Y. Prostaglandin and prostaglandin receptors: present and future promising therapeutic targets for pulmonary arterial hypertension. Respir Res 2023; 24:263. [PMID: 37915044 PMCID: PMC10619262 DOI: 10.1186/s12931-023-02559-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH), Group 1 pulmonary hypertension (PH), is a type of pulmonary vascular disease characterized by abnormal contraction and remodeling of the pulmonary arterioles, manifested by pulmonary vascular resistance (PVR) and increased pulmonary arterial pressure, eventually leading to right heart failure or even death. The mechanisms involved in this process include inflammation, vascular matrix remodeling, endothelial cell apoptosis and proliferation, vasoconstriction, vascular smooth muscle cell proliferation and hypertrophy. In this study, we review the mechanisms of action of prostaglandins and their receptors in PAH. MAIN BODY PAH-targeted therapies, such as endothelin receptor antagonists, phosphodiesterase type 5 inhibitors, activators of soluble guanylate cyclase, prostacyclin, and prostacyclin analogs, improve PVR, mean pulmonary arterial pressure, and the six-minute walk distance, cardiac output and exercise capacity and are licensed for patients with PAH; however, they have not been shown to reduce mortality. Current treatments for PAH primarily focus on inhibiting excessive pulmonary vasoconstriction, however, vascular remodeling is recalcitrant to currently available therapies. Lung transplantation remains the definitive treatment for patients with PAH. Therefore, it is imperative to identify novel targets for improving pulmonary vascular remodeling in PAH. Studies have confirmed that prostaglandins and their receptors play important roles in the occurrence and development of PAH through vasoconstriction, vascular smooth muscle cell proliferation and migration, inflammation, and extracellular matrix remodeling. CONCLUSION Prostacyclin and related drugs have been used in the clinical treatment of PAH. Other prostaglandins also have the potential to treat PAH. This review provides ideas for the treatment of PAH and the discovery of new drug targets.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Jing Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Xialei Zheng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Xinqun Hu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China.
| | - Yuhu He
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China.
| |
Collapse
|
6
|
Guo T, Liu B, Zeng R, Lin R, Guo J, Yu G, Xu Y, Tan X, Xie K, Zhou Y. The vasoconstrictor activities of prostaglandin D 2 via the thromboxane prostanoid receptor and E prostanoid receptor-3 outweigh its concurrent vasodepressor effect mainly through D prostanoid receptor-1 ex vivo and in vivo. Eur J Pharmacol 2023; 956:175963. [PMID: 37543159 DOI: 10.1016/j.ejphar.2023.175963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
Prostaglandin (PG) D2, a commonly considered vasodilator through D prostanoid receptor-1 (DP1), might also evoke vasoconstriction via acting on the thromboxane (Tx)-prostanoid receptor (the original receptor of TxA2; TP) and/or E prostanoid receptor-3 (one of the vasoconstrictor receptors of PGE2; EP3). This study aimed to test the above hypothesis in the mouse renal vascular bed (main renal arteries and perfused kidneys) and/or mesenteric resistance arteries and determine how the vasoconstrictor mechanism influences the overall PGD2 effect on systemic blood pressure under in vivo conditions. Experiments were performed on control wild-type (WT) mice and mice with deficiencies in TP (TP-/-) and/or EP3 (EP3-/-). Here we show that PGD2 indeed evoked vasoconstrictor responses in the above-mentioned tissues of WT mice, which were however not only reduced by TP-/- or EP3-/-, but also reversed by TP-/-/EP3-/- in some of the above tissues (mesenteric resistance arteries or perfused kidneys) to dilator reactions that were reduced by non-selective DP antagonism. A slight or mild pressor response was also observed with PGD2 under in vivo conditions, and this was again reversed to a depressor response in TP-/- or TP-/-/EP3-/- mice. Non-selective DP antagonism reduced the PGD2-evoked depressor response in TP-/-/EP3-/- mice as well. These results thus demonstrate that like other PGs, PGD2 activates TP and/or EP3 to evoke vasoconstrictor activities, which can outweigh its concurrent vasodepressor activity mediated mainly through DP1, and hence result in a pressor response, although the response might only be of a slight or mild extent.
Collapse
Affiliation(s)
- Tingting Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China.
| | - Ruhui Zeng
- Department of Gynaecology and Obstetrics, First Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Rui Lin
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jinwei Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Gang Yu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Yineng Xu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Xiangzhai Tan
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Kaiqi Xie
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China; Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China.
| |
Collapse
|
7
|
Kong D, Yu Y. Prostaglandin D2 signaling and cardiovascular homeostasis. J Mol Cell Cardiol 2022; 167:97-105. [DOI: 10.1016/j.yjmcc.2022.03.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/25/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
8
|
Hrubša M, Siatka T, Nejmanová I, Vopršalová M, Kujovská Krčmová L, Matoušová K, Javorská L, Macáková K, Mercolini L, Remião F, Máťuš M, Mladěnka P, on behalf of the OEMONOM. Biological Properties of Vitamins of the B-Complex, Part 1: Vitamins B 1, B 2, B 3, and B 5. Nutrients 2022; 14:484. [PMID: 35276844 PMCID: PMC8839250 DOI: 10.3390/nu14030484] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
This review summarizes the current knowledge on essential vitamins B1, B2, B3, and B5. These B-complex vitamins must be taken from diet, with the exception of vitamin B3, that can also be synthetized from amino acid tryptophan. All of these vitamins are water soluble, which determines their main properties, namely: they are partly lost when food is washed or boiled since they migrate to the water; the requirement of membrane transporters for their permeation into the cells; and their safety since any excess is rapidly eliminated via the kidney. The therapeutic use of B-complex vitamins is mostly limited to hypovitaminoses or similar conditions, but, as they are generally very safe, they have also been examined in other pathological conditions. Nicotinic acid, a form of vitamin B3, is the only exception because it is a known hypolipidemic agent in gram doses. The article also sums up: (i) the current methods for detection of the vitamins of the B-complex in biological fluids; (ii) the food and other sources of these vitamins including the effect of common processing and storage methods on their content; and (iii) their physiological function.
Collapse
Affiliation(s)
- Marcel Hrubša
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (M.H.); (M.V.); (P.M.)
| | - Tomáš Siatka
- Department of Pharmacognosy, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (T.S.); (K.M.)
| | - Iveta Nejmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Marie Vopršalová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (M.H.); (M.V.); (P.M.)
| | - Lenka Kujovská Krčmová
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic;
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Kralove, Czech Republic; (K.M.); (L.J.)
| | - Kateřina Matoušová
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Kralove, Czech Republic; (K.M.); (L.J.)
| | - Lenka Javorská
- Department of Clinical Biochemistry and Diagnostics, University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Kralove, Czech Republic; (K.M.); (L.J.)
| | - Kateřina Macáková
- Department of Pharmacognosy, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (T.S.); (K.M.)
| | - Laura Mercolini
- Research Group of Pharmaco-Toxicological Analysis (PTA Lab), Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Fernando Remião
- UCIBIO—Applied Molecular Biosciences Unit, REQUINTE, Toxicology Laboratory, Biological Sciences Department Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Marek Máťuš
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovak Republic
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Kralove, Czech Republic; (M.H.); (M.V.); (P.M.)
| | | |
Collapse
|
9
|
Chiang KC, Imig JD, Kalantar-Zadeh K, Gupta A. Kidney in the net of acute and long-haul coronavirus disease 2019: a potential role for lipid mediators in causing renal injury and fibrosis. Curr Opin Nephrol Hypertens 2022; 31:36-46. [PMID: 34846312 DOI: 10.1097/mnh.0000000000000750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Severe COVID-19 disease is often complicated by acute kidney injury (AKI), which may transition to chronic kidney disease (CKD). Better understanding of underlying mechanisms is important in advancing therapeutic approaches. RECENT FINDINGS SARS-CoV-2-induced endothelial injury initiates platelet activation, platelet-neutrophil partnership and release of neutrophil extracellular traps. The resulting thromboinflammation causes ischemia-reperfusion (I/R) injury to end organs. Severe COVID-19 induces a lipid-mediator storm with massive increases in thromboxane A2 (TxA2) and PGD2, which promote thromboinflammation and apoptosis of renal tubular cells, respectively, and thereby enhance renal fibrosis. COVID-19-associated AKI improves rapidly in the majority. However, 15-30% have protracted renal injury, raising the specter of transition from AKI to CKD. SUMMARY In COVID-19, the lipid-mediator storm promotes thromboinflammation, ischemia-reperfusion injury and cytotoxicity. The thromboxane A2 and PGD2 signaling presents a therapeutic target with potential to mitigate AKI and transition to CKD. Ramatroban, the only dual antagonist of the thromboxane A2/TPr and PGD2/DPr2 signaling could potentially mitigate renal injury in acute and long-haul COVID. Urgent studies targeting the lipid-mediator storm are needed to potentially reduce the heavy burden of kidney disease emerging in the wake of the current pandemic.
Collapse
Affiliation(s)
| | - John D Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kamyar Kalantar-Zadeh
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Orange, California, USA
| | - Ajay Gupta
- KARE Biosciences, Orange, California
- Division of Nephrology, Hypertension and Kidney Transplantation, Department of Medicine, University of California Irvine (UCI) School of Medicine, Orange, California, USA
| |
Collapse
|
10
|
Gerges SH, El-Kadi AOS. Sex differences in eicosanoid formation and metabolism: A possible mediator of sex discrepancies in cardiovascular diseases. Pharmacol Ther 2021; 234:108046. [PMID: 34808133 DOI: 10.1016/j.pharmthera.2021.108046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Arachidonic acid is metabolized by cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes to produce prostaglandins, leukotrienes, epoxyeicosatrienoic acids (EETs), and hydroxyeicosatetraenoic acids (HETEs), along with other eicosanoids. Eicosanoids have important physiological and pathological roles in the body, including the cardiovascular system. Evidence from several experimental and clinical studies indicates differences in eicosanoid levels, as well as in the activity or expression levels of their synthesizing and metabolizing enzymes between males and females. In addition, there is a clear state of gender specificity in cardiovascular diseases (CVD), which tend to be more common in men compared to women, and their risk increases significantly in postmenopausal women compared to younger women. This could be largely attributed to sex hormones, as androgens exert detrimental effects on the heart and blood vessels, whereas estrogen exhibits cardioprotective effects. Many of androgen and estrogen effects on the cardiovascular system are mediated by eicosanoids. For example, androgens increase the levels of cardiotoxic eicosanoids like 20-HETE, while estrogens increase the levels of cardioprotective EETs. Thus, sex differences in eicosanoid levels in the cardiovascular system could be an important underlying mechanism for the different effects of sex hormones and the differences in CVD between males and females. Understanding the role of eicosanoids in these differences can help improve the management of CVD.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
11
|
Apolipoprotein B and Cardiovascular Disease: Biomarker and Potential Therapeutic Target. Metabolites 2021; 11:metabo11100690. [PMID: 34677405 PMCID: PMC8540246 DOI: 10.3390/metabo11100690] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein (apo) B, the critical structural protein of the atherogenic lipoproteins, has two major isoforms: apoB48 and apoB100. ApoB48 is found in chylomicrons and chylomicron remnants with one apoB48 molecule per chylomicron particle. Similarly, a single apoB100 molecule is contained per particle of very-low-density lipoprotein (VLDL), intermediate density lipoprotein, LDL and lipoprotein(a). This unique one apoB per particle ratio makes plasma apoB concentration a direct measure of the number of circulating atherogenic lipoproteins. ApoB levels indicate the atherogenic particle concentration independent of the particle cholesterol content, which is variable. While LDL, the major cholesterol-carrying serum lipoprotein, is the primary therapeutic target for management and prevention of atherosclerotic cardiovascular disease, there is strong evidence that apoB is a more accurate indicator of cardiovascular risk than either total cholesterol or LDL cholesterol. This review examines multiple aspects of apoB structure and function, with a focus on the controversy over use of apoB as a therapeutic target in clinical practice. Ongoing coronary artery disease residual risk, despite lipid-lowering treatment, has left patients and clinicians with unsatisfactory options for monitoring cardiovascular health. At the present time, the substitution of apoB for LDL-C in cardiovascular disease prevention guidelines has been deemed unjustified, but discussions continue.
Collapse
|
12
|
Säfholm J, Abma W, Liu J, Balgoma D, Fauland A, Kolmert J, Wheelock CE, Adner M, Dahlén SE. Prostaglandin D 2 inhibits mediator release and antigen induced bronchoconstriction in the Guinea pig trachea by activation of DP 1 receptors. Eur J Pharmacol 2021; 907:174282. [PMID: 34175307 DOI: 10.1016/j.ejphar.2021.174282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
The mechanism by which cyclooxygenase (COX) inhibition increases antigen-induced responses in airways remains unknown. Male albino guinea pigs were sensitized to ovalbumin (OVA). Intact rings of the trachea were isolated and mounted in organ baths for either force measurements or lipid mediator release analysis by UPLC-MS/MS or EIA following relevant pharmacological interventions. First, challenge with OVA increased the release of all primary prostanoids (prostaglandin (PG) D2/E2/F2α/I2 and thromboxanes). This release was eliminated by unselective COX inhibition (indomethacin) whereas selective inhibition of COX-2 (lumiracoxib) did not inhibit release of PGD2 or thromboxanes. Additionally, the increased levels of leukotriene B4 and E4 after OVA were further amplified by unselective COX inhibition. Second, unselective inhibition of COX and selective inhibition of the prostaglandin D synthase (2-Phenyl-Pyrimidine-5-Carboxylic Acid (2,3-dihydro-indol-1-yl)-amide) amplified the antigen-induced bronchoconstriction which was reversed by exogenous PGD2. Third, a DP1 receptor agonist (BW 245c) concentration-dependently reduced the antigen-induced constriction as well as reducing released histamine and cysteinyl-leukotrienes, a response inhibited by the DP1 receptor antagonist (MK-524). In contrast, a DP2 receptor agonist (15(R)-15-methyl PGD2) failed to modulate the OVA-induced constriction. In the guinea pig trachea, endogenous PGD2 is generated via COX-1 and mediates an inhibitory effect of the antigen-induced bronchoconstriction via DP1 receptors inhibiting mast cell release of bronchoconstrictive mediators. Removal of this protective function by COX-inhibition results in increased release of mast cell mediators and enhanced bronchoconstriction.
Collapse
Affiliation(s)
- Jesper Säfholm
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.
| | - Willem Abma
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Jielu Liu
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - David Balgoma
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Alexander Fauland
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Kolmert
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden; Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Adner
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
In situ imaging reveals disparity between prostaglandin localization and abundance of prostaglandin synthases. Commun Biol 2021; 4:966. [PMID: 34389796 PMCID: PMC8363604 DOI: 10.1038/s42003-021-02488-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 07/22/2021] [Indexed: 12/14/2022] Open
Abstract
Prostaglandins are important lipids involved in mediating many physiological processes, such as allergic responses, inflammation, and pregnancy. However, technical limitations of in-situ prostaglandin detection in tissue have led researchers to infer prostaglandin tissue distributions from localization of regulatory synthases, such as COX1 and COX2. Herein, we apply a novel mass spectrometry imaging method for direct in situ tissue localization of prostaglandins, and combine it with techniques for protein expression and RNA localization. We report that prostaglandin D2, its precursors, and downstream synthases co-localize with the highest expression of COX1, and not COX2. Further, we study tissue with a conditional deletion of transformation-related protein 53 where pregnancy success is low and confirm that PG levels are altered, although localization is conserved. Our studies reveal that the abundance of COX and prostaglandin D2 synthases in cellular regions does not mirror the regional abundance of prostaglandins. Thus, we deduce that prostaglandins tissue localization and abundance may not be inferred by COX or prostaglandin synthases in uterine tissue, and must be resolved by an in situ prostaglandin imaging. Duncan et al. use a mass spectrometry imaging method to assess the localization and concentration of prostaglandins (PGs) in mouse tissues during pregnancy. This study brings new biological insights into the spatial evaluation of PGs in tissues, which could reveal the functional significance of each PGs during different stages of embryo development/pregnancy.
Collapse
|
14
|
Buchheit KM, Lewis E, Gakpo D, Hacker J, Sohail A, Taliaferro F, Berreondo Giron E, Asare C, Vukovic M, Bensko JC, Dwyer DF, Shalek AK, Ordovas-Montanes J, Laidlaw TM. Mepolizumab targets multiple immune cells in aspirin-exacerbated respiratory disease. J Allergy Clin Immunol 2021; 148:574-584. [PMID: 34144111 PMCID: PMC9096876 DOI: 10.1016/j.jaci.2021.05.043] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Eosinophilic asthma and nasal polyposis are hallmarks of aspirin-exacerbated respiratory disease (AERD), and IL-5 inhibition has been shown to provide therapeutic benefit. However, IL-5Rα is expressed on many cells in addition to eosinophils, and the mechanisms by which IL-5 inhibition leads to clinical benefit in eosinophilic asthma and nasal polyposis are unlikely to be due exclusively to antieosinophil effects. OBJECTIVE We sought to identify the mechanisms by which anti-IL-5 treatment with mepolizumab improves respiratory inflammation in AERD. METHODS The clinical characteristics, circulating granulocytes, nasal scraping transcripts, eosinophilic cationic protein, tryptase, and antibody levels, and urinary and nasal eicosanoid levels were measured for 18 subjects with AERD who were taking mepolizumab and compared with those of 18 matched subjects with AERD who were not taking mepolizumab. RESULTS Subjects taking mepolizumab had significantly fewer peripheral blood eosinophils and basophils, and those cells that remained had higher surface CRTH2 expression than did the cells from subjects not taking mepolizumab. Nasal prostaglandin F2α, prostaglandin D2 metabolites, leukotriene B4, and thromboxane levels were lower in subjects taking mepolizumab, as were urinary levels of tetranor-prostaglandin D2 and leukotriene E4. The nasal epithelial cell transcripts that were overexpressed among subjects with AERD who were taking mepolizumab were enriched for genes involved in tight junction formation and cilium organization. Nasal and urinary prostaglandin E2, tryptase, and antibody levels were not different between the 2 groups. CONCLUSION IL-5 inhibition in AERD decreases production of inflammatory eicosanoids and upregulates tight junction-associated nasal epithelial cell transcripts, likely due to decreased IL-5 signaling on tissue mast cells, eosinophils, and epithelial cells. These direct effects on multiple relevant immune cells contribute to the mechanism of benefit afforded by mepolizumab.
Collapse
Affiliation(s)
- Kathleen M Buchheit
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Erin Lewis
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Deborah Gakpo
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Jonathan Hacker
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Aaqib Sohail
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Faith Taliaferro
- Division of Gastroenterology, Boston Children's Hospital, Boston, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass
| | | | - Chelsea Asare
- Division of Gastroenterology, Boston Children's Hospital, Boston, Mass
| | - Marko Vukovic
- Broad Institute of MIT and Harvard, Cambridge, Mass; Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, Mass; Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, Mass
| | - Jillian C Bensko
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Daniel F Dwyer
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, Mass; Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, Mass; Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, Mass; Program in Immunology, Harvard Medical School, Boston, Mass; Harvard-MIT Division of Health Sciences & Technology, Cambridge, Mass
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Boston Children's Hospital, Boston, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass; Program in Immunology, Harvard Medical School, Boston, Mass; Harvard Stem Cell Institute, Cambridge, Mass
| | - Tanya M Laidlaw
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass.
| |
Collapse
|
15
|
Alfaro GF, Rodriguez-Zas SL, Southey BR, Muntifering RB, Rodning SP, Pacheco WJ, Moisá SJ. Complete Blood Count Analysis on Beef Cattle Exposed to Fescue Toxicity and Rumen-Protected Niacin Supplementation. Animals (Basel) 2021; 11:ani11040988. [PMID: 33916070 PMCID: PMC8065407 DOI: 10.3390/ani11040988] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Fescue toxicity results from cattle consuming fungal (Ergot spp.) endophyte-infected tall fescue. Ergot alkaloids like ergovaline produce vasoconstriction in cattle. Our objectives were to analyze changes in complete blood count and performance due to ergot alkaloid detoxification in growing beef cattle and the effect of selecting dams tolerant or susceptible to fescue toxicity based on their tolerance index, measured through a genetic test currently available for beef producers. Furthermore, rumen-protected niacin supplementation is proposed as a potential alleviator for vasoconstriction produced by fescue toxicity. Therefore, we assessed the effects of consuming endophyte-infected tall fescue seeds in addition to rumen-protected niacin supplementation in offspring performance and hematological parameters. Signs for anemia were noticed in susceptible heifer offspring that did not receive rumen-protected niacin, whereas inflammation or infection was detected in tolerant steers that received niacin in their diet. Typical symptoms of heat stress and intoxication with ergot alkaloids were noticed in offspring. Our results suggest that susceptible heifer offspring might have a more active detox metabolism when under fescue toxicity. Findings from this study could be utilized as a new tool to help beef cattle producers to dampen the adverse effects of fescue toxicity. Abstract Offspring born to dams genetically tested for resistance to fescue toxicity were separated in groups based on their dams’ resistance level (tolerant vs. susceptible). Rumen-protected niacin (RPN) is proposed as a potential alleviator for vasoconstriction produced by fescue toxicity. Complete blood count (CBC) analysis was utilized for detection of significant responses to treatments applied. Our objectives were as follows: (a) to analyze changes in CBC due to fescue toxicity, maternal resistance level, and RPN in growing offspring; and (b) to assess the effects of maternal resistance level when consuming endophyte-infected tall fescue seeds in addition to RPN in offspring performance. Body weight, average daily gain, or health status were not improved by RPN or the genetic test to detect fescue toxicity resistance. Typical signs of alkaloids intoxication and heat stress were noticed in offspring. Particularly, rectal temperature was greater for susceptible control heifers. Results showed that susceptible control offspring presented signs of anemia denoted by low mean corpuscular hemoglobin (MCH) and mean corpuscular volume (MCV). High levels of white blood cells (WBC) and basophils in combination to low neutrophils to lymphocytes ratio were the signs of infection or inflammation detected in the CBC analysis, especially in tolerant niacin steers. Furthermore, offspring of control heifers had a greater percentage of reticulocytes and RDW, denoting signs of anemia.
Collapse
Affiliation(s)
- Gaston F. Alfaro
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (G.F.A.); (R.B.M.); (S.P.R.)
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA; (S.L.R.-Z.); (B.R.S.)
| | - Bruce R. Southey
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA; (S.L.R.-Z.); (B.R.S.)
| | - Russell B. Muntifering
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (G.F.A.); (R.B.M.); (S.P.R.)
| | - Soren P. Rodning
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (G.F.A.); (R.B.M.); (S.P.R.)
| | - Wilmer J. Pacheco
- Department of Poultry Sciences, Auburn University, Auburn, AL 36849, USA;
| | - Sonia J. Moisá
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (G.F.A.); (R.B.M.); (S.P.R.)
- Correspondence:
| |
Collapse
|
16
|
Zhao S, Cheng CK, Zhang CL, Huang Y. Interplay Between Oxidative Stress, Cyclooxygenases, and Prostanoids in Cardiovascular Diseases. Antioxid Redox Signal 2021; 34:784-799. [PMID: 32323554 DOI: 10.1089/ars.2020.8105] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Significance: Endothelial cells lining the lumen of blood vessels play an important role in the regulation of cardiovascular functions through releasing both vasoconstricting and vasodilating factors. The production and function of vasoconstricting factors are largely elevated in hypertension, diabetes, atherosclerosis, and ischemia/reperfusion injuries. Cyclooxygenases (COXs) are the major enzymes producing five different prostanoids that act as either contracting or relaxing substances. Under conditions of increased oxidative stress, the expressions and activities of COX isoforms are altered, resulting in changes in production of various prostanoids and thus affecting vascular tone. This review briefly summarizes the relationship between oxidative stress, COXs, and prostanoids, thereby providing new insights into the pathophysiological mechanisms of cardiovascular diseases (CVDs). Recent Advances: Many new drugs targeting oxidative stress, COX-2, and prostanoids against common CVDs have been evaluated in recent years and they are summarized in this review. Critical Issues: Comprehensive understanding of the complex interplay between oxidative stress, COXs, and prostanoids in CVDs helps develop more effective measures against cardiovascular pathogenesis. Future Directions: Apart from minimizing the undesired effects of harmful prostanoids, future studies shall investigate the restoration of vasoprotective prostanoids as a means to combat CVDs. Antioxid. Redox Signal. 34, 784-799.
Collapse
Affiliation(s)
- Sha Zhao
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chak Kwong Cheng
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Cheng-Lin Zhang
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Heart and Vascular Institute and School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Gene expression of the heat stress response in bovine peripheral white blood cells and milk somatic cells in vivo. Sci Rep 2020; 10:19181. [PMID: 33154392 PMCID: PMC7645416 DOI: 10.1038/s41598-020-75438-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
Abstract
Heat stress in dairy cattle leads to reduction in feed intake and milk production as well as the induction of many physiological stress responses. The genes implicated in the response to heat stress in vivo are not well characterised. With the aim of identifying such genes, an experiment was conducted to perform differential gene expression in peripheral white blood cells and milk somatic cells in vivo in 6 Holstein Friesian cows in thermoneutral conditions and in 6 Holstein Friesian cows exposed to a short-term moderate heat challenge. RNA sequences from peripheral white blood cells and milk somatic cells were used to quantify full transcriptome gene expression. Genes commonly differentially expressed (DE) in both the peripheral white blood cells and in milk somatic cells were associated with the cellular stress response, apoptosis, oxidative stress and glucose metabolism. Genes DE in peripheral white blood cells of cows exposed to the heat challenge compared to the thermoneutral control were related to inflammation, lipid metabolism, carbohydrate metabolism and the cardiovascular system. Genes DE in milk somatic cells compared to the thermoneutral control were involved in the response to stress, thermoregulation and vasodilation. These findings provide new insights into the cellular adaptations induced during the response to short term moderate heat stress in dairy cattle and identify potential candidate genes (BDKRB1 and SNORA19) for future research.
Collapse
|
18
|
Jia D, Bai P, Wan N, Liu J, Zhu Q, He Y, Chen G, Wang J, Chen H, Wang C, Lyu A, Lazarus M, Su Y, Urade Y, Yu Y, Zhang J, Shen Y. Niacin Attenuates Pulmonary Hypertension Through H-PGDS in Macrophages. Circ Res 2020; 127:1323-1336. [PMID: 32912104 DOI: 10.1161/circresaha.120.316784] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is characterized by progressive pulmonary vascular remodeling, accompanied by varying degrees of perivascular inflammation. Niacin, a commonly used lipid-lowering drug, possesses vasodilating and proresolution effects by promoting the release of prostaglandin D2 (PGD2). However, whether or not niacin confers protection against PAH pathogenesis is still unknown. OBJECTIVE This study aimed to determine whether or not niacin attenuates the development of PAH and, if so, to elucidate the molecular mechanisms underlying its effects. METHODS AND RESULTS Vascular endothelial growth factor receptor inhibitor SU5416 and hypoxic exposure were used to induce pulmonary hypertension (PH) in rodents. We found that niacin attenuated the development of this hypoxia/SU5416-induced PH in mice and suppressed progression of monocrotaline-induced and hypoxia/SU5416-induced PH in rats through the reduction of pulmonary artery remodeling. Niacin boosted PGD2 generation in lung tissue, mainly through H-PGDS (hematopoietic PGD2 synthases). Deletion of H-PGDS, but not lipocalin-type PGDS, exacerbated the hypoxia/SU5416-induced PH in mice and abolished the protective effects of niacin against PAH. Moreover, H-PGDS was expressed dominantly in infiltrated macrophages in lungs of PH mice and patients with idiopathic PAH. Macrophage-specific deletion of H-PGDS markedly decreased PGD2 generation in lungs, aggravated hypoxia/SU5416-induced PH in mice, and attenuated the therapeutic effect of niacin on PAH. CONCLUSIONS Niacin treatment ameliorates the progression of PAH through the suppression of vascular remodeling by stimulating H-PGDS-derived PGD2 release from macrophages.
Collapse
Affiliation(s)
- Daile Jia
- Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.J., J.L., G.C., Y.Y., J.Z., Y. Shen).,Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China (D.J., P.B.).,Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (D.J., P.B., N.W., Q.Z., Y.H., Y.Y.)
| | - Peiyuan Bai
- Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China (D.J., P.B.).,Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (D.J., P.B., N.W., Q.Z., Y.H., Y.Y.)
| | - Naifu Wan
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (D.J., P.B., N.W., Q.Z., Y.H., Y.Y.).,Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (N.W., Q.Z., A.L.)
| | - Jiao Liu
- Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.J., J.L., G.C., Y.Y., J.Z., Y. Shen).,Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (J.L., Y.Y.)
| | - Qian Zhu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (D.J., P.B., N.W., Q.Z., Y.H., Y.Y.).,Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (N.W., Q.Z., A.L.)
| | - Yuhu He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (D.J., P.B., N.W., Q.Z., Y.H., Y.Y.)
| | - Guilin Chen
- Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.J., J.L., G.C., Y.Y., J.Z., Y. Shen)
| | - Jing Wang
- Cardiology, Cardiovascular Institute and Fuwai Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China (J.W.)
| | - Han Chen
- Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China (H.C., C.W.)
| | - Chen Wang
- Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China (H.C., C.W.)
| | - Ankang Lyu
- Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (N.W., Q.Z., A.L.)
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba City, Japan (M.L.)
| | - Yunchao Su
- Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Georgia, United States of America (Y. Su)
| | - Yoshihiro Urade
- Isotope Science Center, The University of Tokyo, Tokyo, Japan (Y.U.)
| | - Ying Yu
- Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.J., J.L., G.C., Y.Y., J.Z., Y. Shen).,Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (D.J., P.B., N.W., Q.Z., Y.H., Y.Y.).,Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (J.L., Y.Y.)
| | - Jian Zhang
- Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.J., J.L., G.C., Y.Y., J.Z., Y. Shen)
| | - Yujun Shen
- Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.J., J.L., G.C., Y.Y., J.Z., Y. Shen)
| |
Collapse
|
19
|
He Y, Zuo C, Jia D, Bai P, Kong D, Chen D, Liu G, Li J, Wang Y, Chen G, Yan S, Xiao B, Zhang J, Piao L, Li Y, Deng Y, Li B, Roux PP, Andreasson KI, Breyer RM, Su Y, Wang J, Lyu A, Shen Y, Yu Y. Loss of DP1 Aggravates Vascular Remodeling in Pulmonary Arterial Hypertension via mTORC1 Signaling. Am J Respir Crit Care Med 2020; 201:1263-1276. [PMID: 31917615 DOI: 10.1164/rccm.201911-2137oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Rationale: Vascular remodeling, including smooth muscle cell hypertrophy and proliferation, is the key pathological feature of pulmonary arterial hypertension (PAH). Prostaglandin I2 analogs (beraprost, iloprost, and treprostinil) are effective in the treatment of PAH. Of note, the clinically favorable effects of treprostinil in severe PAH may be attributable to concomitant activation of DP1 (D prostanoid receptor subtype 1).Objectives: To study the role of DP1 in the progression of PAH and its underlying mechanism.Methods: DP1 levels were examined in pulmonary arteries of patients and animals with PAH. Multiple genetic and pharmacologic approaches were used to investigate DP1-mediated signaling in PAH.Measurements and Main Results: DP1 expression was downregulated in hypoxia-treated pulmonary artery smooth muscle cells and in pulmonary arteries from rodent PAH models and patients with idiopathic PAH. DP1 deletion exacerbated pulmonary artery remodeling in hypoxia-induced PAH, whereas pharmacological activation or forced expression of the DP1 receptor had the opposite effect in different rodent models. DP1 deficiency promoted pulmonary artery smooth muscle cell hypertrophy and proliferation in response to hypoxia via induction of mTORC1 (mammalian target of rapamycin complex 1) activity. Rapamycin, an inhibitor of mTORC1, alleviated the hypoxia-induced exacerbation of PAH in DP1-knockout mice. DP1 activation facilitated raptor dissociation from mTORC1 and suppressed mTORC1 activity through PKA (protein kinase A)-dependent phosphorylation of raptor at Ser791. Moreover, treprostinil treatment blocked the progression of hypoxia-induced PAH in mice in part by targeting the DP1 receptor.Conclusions: DP1 activation attenuates hypoxia-induced pulmonary artery remodeling and PAH through PKA-mediated dissociation of raptor from mTORC1. These results suggest that the DP1 receptor may serve as a therapeutic target for the management of PAH.
Collapse
Affiliation(s)
- Yuhu He
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Department of Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Caojian Zuo
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Department of Cardiology, Shanghai General Hospital, and
| | - Daile Jia
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peiyuan Bai
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Deping Kong
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Di Chen
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guizhu Liu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Juanjuan Li
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyang Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Guilin Chen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shuai Yan
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bing Xiao
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jian Zhang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lingjuan Piao
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanli Li
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yi Deng
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bin Li
- Orthopedic Institute, Soochow University, Jiangsu, China
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer and.,Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, Tennessee.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ankang Lyu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yujun Shen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Jara CP, Mendes NF, Prado TPD, de Araújo EP. Bioactive Fatty Acids in the Resolution of Chronic Inflammation in Skin Wounds. Adv Wound Care (New Rochelle) 2020; 9:472-490. [PMID: 32320357 DOI: 10.1089/wound.2019.1105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Significance: Optimal skin wound healing is crucial for maintaining tissue homeostasis, particularly in response to an injury. The skin immune system is under regulation of mediators such as bioactive lipids and cytokines that can initiate an immune response with controlled inflammation, followed by efficient resolution. However, nutritional deficiency impacts wound healing by hindering fibroblast proliferation, collagen synthesis, and epithelialization, among other crucial functions. In this way, the correct nutritional support of bioactive lipids and of other essential nutrients plays an important role in the outcome of the wound healing process. Recent Advances and Critical Issues: Several studies have revealed the potential role of lipids as a treatment for the healing of skin wounds. Unsaturated fatty acids such as linoleic acid, α-linolenic acid, oleic acid, and most of their bioactive products have shown an effective role as a topical treatment of chronic skin wounds. Their effect, when the treatment starts at day 0, has been observed mainly in the inflammatory phase of the wound healing process. Moreover, some of them were associated with different dressings and were tested for clinical purposes, including pluronic gel, nanocapsules, collagen films and matrices, and polymeric bandages. Therefore, future research is still needed to evaluate these dressing technologies in association with different bioactive fatty acids in a wound healing context. Future Directions: This review summarizes the main results of the available clinical trials and basic research studies and provides evidence-based conclusions. Together, current data encourage the use of bioactive fatty acids for an optimal wound healing resolution.
Collapse
Affiliation(s)
- Carlos Poblete Jara
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Natália Ferreira Mendes
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Thais Paulino do Prado
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Eliana Pereira de Araújo
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
21
|
Zhu L, Zhang Y, Guo Z, Wang M. Cardiovascular Biology of Prostanoids and Drug Discovery. Arterioscler Thromb Vasc Biol 2020; 40:1454-1463. [PMID: 32295420 DOI: 10.1161/atvbaha.119.313234] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Prostanoids are a group of bioactive lipids that are synthesized de novo from membrane phospholipid-released arachidonic acid and have diverse functions in normal physiology and disease. NSAIDs (non-steroidal anti-inflammatory drugs), which are among the most commonly used medications, ameliorate pain, fever, and inflammation by inhibiting COX (cyclooxygenase), which is the rate-limiting enzyme in the biosynthetic cascade of prostanoids. The use of NSAIDs selective for COX-2 inhibition increases the risk of a thrombotic event (eg, myocardial infarction and stroke). All NSAIDs are associated with an increased risk of heart failure. Substantial variation in clinical responses to aspirin exists and is associated with cardiovascular risk. Limited clinical studies suggest the involvement of prostanoids in vascular restenosis in patients who received angioplasty intervention. mPGES (microsomal PG [prostaglandin] E synthase)-1, an alternative target downstream of COX, has the potential to be therapeutically targeted for inflammatory disease, with diminished thrombotic risk relative to selective COX-2 inhibitors. mPGES-1-derived PGE2 critically regulates microcirculation via its receptor EP (receptor for prostanoid E) 4. This review summarizes the actions and associated mechanisms for modulating the biosynthesis of prostanoids in thrombosis, vascular remodeling, and ischemic heart disease as well as their therapeutic relevance.
Collapse
Affiliation(s)
- Liyuan Zhu
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Yuze Zhang
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Ziyi Guo
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Miao Wang
- From the State Key Laboratory of Cardiovascular Disease (L.Z., Y.Z., Z.G., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing.,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| |
Collapse
|
22
|
Brightling CE, Brusselle G, Altman P. The impact of the prostaglandin D 2 receptor 2 and its downstream effects on the pathophysiology of asthma. Allergy 2020; 75:761-768. [PMID: 31355946 DOI: 10.1111/all.14001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/24/2019] [Accepted: 07/17/2019] [Indexed: 02/03/2023]
Abstract
Current research suggests that the prostaglandin D2 (PGD2 ) receptor 2 (DP2 ) is a principal regulator in the pathophysiology of asthma, because it stimulates and amplifies the inflammatory response in this condition. The DP2 receptor can be activated by both allergic and nonallergic stimuli, leading to several pro-inflammatory events, including eosinophil activation and migration, release of the type 2 cytokines interleukin (IL)-4, IL-5 and IL-13 from T helper 2 (Th2) cells and innate lymphoid cells type 2 (ILCs), and increased airway smooth muscle mass via recruitment of mesenchymal progenitors to the airway smooth muscle bundle. Activation of the DP2 receptor pathway has potential downstream effects on asthma pathophysiology, including on airway epithelial cells, mucus hypersecretion, and airway remodelling, and consequently might impact asthma symptoms and exacerbations. Given the broad distribution of DP2 receptors on immune and structural cells involved in asthma, this receptor is being explored as a novel therapeutic target.
Collapse
Affiliation(s)
| | - Guy Brusselle
- Department of Respiratory Diseases Ghent University Hospital Ghent Belgium
| | - Pablo Altman
- Novartis Pharmaceuticals Corporation East Hanover NJ USA
| |
Collapse
|
23
|
Davidsson Ö, Nilsson K, Brånalt J, Andersson T, Berggren K, Chen Y, Fjellström O, Gradén H, Gustafsson L, Hermansson NO, Jansen F, Johannesson P, Ohlsson B, Tyrchan C, Wellner A, Wellner E, Ölwegård-Halvarsson M. Identification of novel GPR81 agonist lead series for target biology evaluation. Bioorg Med Chem Lett 2020; 30:126953. [PMID: 31932225 DOI: 10.1016/j.bmcl.2020.126953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/01/2020] [Indexed: 02/07/2023]
Abstract
GPR81 is a novel drug target that is implicated in the control of glucose and lipid metabolism. The lack of potent GPR81 modulators suitable for in vivo studies has limited the pharmacological characterization of this lactate sensing receptor. We performed a high throughput screen (HTS) and identified a GPR81 agonist chemical series containing a central acyl urea scaffold linker. During SAR exploration two additional new series were evolved, one containing cyclic acyl urea bioisosteres and another a central amide bond. These three series provide different selectivity and physicochemical properties suitable for in-vivo studies.
Collapse
Affiliation(s)
- Öjvind Davidsson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Kristina Nilsson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Jonas Brånalt
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Terese Andersson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Product Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kristina Berggren
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; RIA Medicinal Chemistry, Research and Early Development, Respiratory, Inflammation and Autoimmune, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yantao Chen
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ola Fjellström
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Henrik Gradén
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda Gustafsson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Frank Jansen
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Petra Johannesson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Global Patient Safety CVRM, Chief Medical Office, AstraZeneca R&D, Gothenburg, Sweden
| | - Bengt Ohlsson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christian Tyrchan
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; RIA Medicinal Chemistry, Research and Early Development, Respiratory, Inflammation and Autoimmune, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Annika Wellner
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; RIA Medicinal Chemistry, Research and Early Development, Respiratory, Inflammation and Autoimmune, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eric Wellner
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Ölwegård-Halvarsson
- CVRM Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
24
|
Szentirmai É, Kapás L. Nicotinic acid promotes sleep through prostaglandin synthesis in mice. Sci Rep 2019; 9:17084. [PMID: 31745228 PMCID: PMC6863905 DOI: 10.1038/s41598-019-53648-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/01/2019] [Indexed: 12/26/2022] Open
Abstract
Nicotinic acid has been used for decades for its antiatherogenic properties in humans. Its actions on lipid metabolism intersect with multiple sleep regulatory mechanisms, but its effects on sleep have never been documented. For the first time, we investigated the effects of acute systemic administration of nicotinic acid on sleep in mice. Intraperitoneal and oral gavage administration of nicotinic acid elicited robust increases in non-rapid-eye movement sleep (NREMS) and decreases in body temperature, energy expenditure and food intake. Preventing hypothermia did not affect its sleep-inducing actions suggesting that altered sleep is not secondary to decreased body temperature. Systemic administration of nicotinamide, a conversion product of nicotinic acid, did not affect sleep amounts and body temperature, indicating that it is not nicotinamide that underlies these actions. Systemic administration of monomethyl fumarate, another agonist of the nicotinic acid receptor GPR109A, fully recapitulated the somnogenic and thermoregulatory effects of nicotinic acid suggesting that they are mediated by the GPR109A receptor. The cyclooxygenase inhibitor indomethacin completely abolished the effects of nicotinic acid indicating that prostaglandins play a key role in mediating the sleep and thermoregulatory responses of nicotinic acid.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America.
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America.
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America
| |
Collapse
|
25
|
Werder RB, Lynch JP, Simpson JC, Zhang V, Hodge NH, Poh M, Forbes-Blom E, Kulis C, Smythe ML, Upham JW, Spann K, Everard ML, Phipps S. PGD2/DP2 receptor activation promotes severe viral bronchiolitis by suppressing IFN- λ production. Sci Transl Med 2019; 10:10/440/eaao0052. [PMID: 29743346 DOI: 10.1126/scitranslmed.aao0052] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 11/17/2017] [Accepted: 03/12/2018] [Indexed: 12/27/2022]
Abstract
Prostaglandin D2 (PGD2) signals through PGD2 receptor 2 (DP2, also known as CRTH2) on type 2 effector cells to promote asthma pathogenesis; however, little is known about its role during respiratory syncytial virus (RSV) bronchiolitis, a major risk factor for asthma development. We show that RSV infection up-regulated hematopoietic prostaglandin D synthase expression and increased PGD2 release by cultured human primary airway epithelial cells (AECs). Moreover, PGD2 production was elevated in nasopharyngeal samples from young infants hospitalized with RSV bronchiolitis compared to healthy controls. In a neonatal mouse model of severe viral bronchiolitis, DP2 antagonism decreased viral load, immunopathology, and morbidity and ablated the predisposition for subsequent asthma onset in later life. This protective response was abolished upon dual DP1/DP2 antagonism and replicated with a specific DP1 agonist. Rather than mediating an effect via type 2 inflammation, the beneficial effects of DP2 blockade or DP1 agonism were associated with increased interferon-λ (IFN-λ) [interleukin-28A/B (IL-28A/B)] expression and were lost upon IL-28A neutralization. In RSV-infected AEC cultures, DP1 activation up-regulated IFN-λ production, which, in turn, increased IFN-stimulated gene expression, accelerating viral clearance. Our findings suggest that DP2 antagonists or DP1 agonists may be useful antivirals for the treatment of viral bronchiolitis and possibly as primary preventatives for asthma.
Collapse
Affiliation(s)
- Rhiannon B Werder
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
| | - Jason P Lynch
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
| | - Jennifer C Simpson
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia.,Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston 4006, Australia
| | - Vivian Zhang
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston 4006, Australia
| | - Nick H Hodge
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
| | - Matthew Poh
- School of Paediatrics and Child Health, University of Western Australia, Western Australia 6840, Australia
| | | | - Christina Kulis
- Institute for Molecular Bioscience, University of Queensland, Queensland 4072, Australia
| | - Mark L Smythe
- Institute for Molecular Bioscience, University of Queensland, Queensland 4072, Australia
| | - John W Upham
- Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland 4102, Australia
| | - Kirsten Spann
- Australian Infectious Diseases Research Centre, University of Queensland, Queensland 4067, Australia.,School of Biomedical Sciences, Queensland University of Technology, Queensland 4001, Australia
| | - Mark L Everard
- School of Paediatrics and Child Health, University of Western Australia, Western Australia 6840, Australia
| | - Simon Phipps
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston 4006, Australia. .,Australian Infectious Diseases Research Centre, University of Queensland, Queensland 4067, Australia
| |
Collapse
|
26
|
Min L, Li D, Tong X, Nan X, Ding D, Xu B, Wang G. Nutritional strategies for alleviating the detrimental effects of heat stress in dairy cows: a review. INTERNATIONAL JOURNAL OF BIOMETEOROLOGY 2019; 63:1283-1302. [PMID: 31218396 DOI: 10.1007/s00484-019-01744-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 03/29/2019] [Accepted: 06/05/2019] [Indexed: 06/09/2023]
Abstract
Heat stress responses negatively impact production performance, milk quality, body temperature, and other parameters in dairy cows. As global warming continues unabated, heat stress in dairy cows is likely to become more widespread in the future. To address this challenge, researchers have evaluated a number of potentially available nutritional strategies, including dietary fat, dietary fiber, dietary microbial additives, minerals, vitamins, metal ion buffer, plant extracts, and other anti-stress additives. In this paper, we discuss the evidence for the efficacy of these nutritional strategies aimed at alleviating the detrimental effects of heat stress in dairy cows. It was comprised of the treatment (dosage and usage), animal information (lactation stage and number of dairy cows), THI value (level of heat stress), duration of exposure, the changes of feed intake and milk yield (production performance), the changes of milk protein and milk fat (milk quality), the changes of rectal temperature and respiration rate (body temperature), other indices, and reference resources. The results of these studies are presented with statistical justification in the tables. In total, the 49 kinds of dietary interventions derived from these eight types of nutritional strategies may provide an appropriate means of mitigating heat stress on a particular dairy farm based on the explanation of the results.
Collapse
Affiliation(s)
- Li Min
- State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
| | - Dagang Li
- State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
| | - Xiong Tong
- State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Diyun Ding
- State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
| | - Bin Xu
- State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China
| | - Gang Wang
- State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, People's Republic of China.
| |
Collapse
|
27
|
Ahmad AS, Ottallah H, Maciel CB, Strickland M, Doré S. Role of the L-PGDS-PGD2-DP1 receptor axis in sleep regulation and neurologic outcomes. Sleep 2019; 42:zsz073. [PMID: 30893431 PMCID: PMC6559173 DOI: 10.1093/sleep/zsz073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/16/2019] [Indexed: 12/18/2022] Open
Abstract
To meet the new challenges of modern lifestyles, we often compromise a good night's sleep. In preclinical models as well as in humans, a chronic lack of sleep is reported to be among the leading causes of various physiologic, psychologic, and neurocognitive deficits. Thus far, various endogenous mediators have been implicated in inter-regulatory networks that collectively influence the sleep-wake cycle. One such mediator is the lipocalin-type prostaglandin D2 synthase (L-PGDS)-Prostaglandin D2 (PGD2)-DP1 receptor (L-PGDS-PGD2-DP1R) axis. Findings in preclinical models confirm that DP1R are predominantly expressed in the sleep-regulating centers. This finding led to the discovery that the L-PGDS-PGD2-DP1R axis is involved in sleep regulation. Furthermore, we showed that the L-PGDS-PGD2-DP1R axis is beneficial in protecting the brain from ischemic stroke. Protein sequence homology was also performed, and it was found that L-PGDS and DP1R share a high degree of homology between humans and rodents. Based on the preclinical and clinical data thus far pertaining to the role of the L-PGDS-PGD2-DP1R axis in sleep regulation and neurologic conditions, there is optimism that this axis may have a high translational potential in human therapeutics. Therefore, here the focus is to review the regulation of the homeostatic component of the sleep process with a special focus on the L-PGDS-PGD2-DP1R axis and the consequences of sleep deprivation on health outcomes. Furthermore, we discuss whether the pharmacological regulation of this axis could represent a tool to prevent sleep disturbances and potentially improve outcomes, especially in patients with acute brain injuries.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Haneen Ottallah
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Carolina B Maciel
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL
| | - Michael Strickland
- Division of Biology and Biomedical Sciences, Washington University in Saint Louis, Saint Louis, MO
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
- Department of Psychiatry, University of Florida, Gainesville, FL
- Department of Pharmaceutics, University of Florida, Gainesville, FL
- Department of Psychology, University of Florida, Gainesville, FL
- Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
28
|
Wang TA, Teo CF, Åkerblom M, Chen C, Tynan-La Fontaine M, Greiner VJ, Diaz A, McManus MT, Jan YN, Jan LY. Thermoregulation via Temperature-Dependent PGD 2 Production in Mouse Preoptic Area. Neuron 2019; 103:309-322.e7. [PMID: 31151773 DOI: 10.1016/j.neuron.2019.04.035] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 04/03/2019] [Accepted: 04/24/2019] [Indexed: 12/30/2022]
Abstract
Body temperature control is essential for survival. In mammals, thermoregulation is mediated by the preoptic area of anterior hypothalamus (POA), with ∼30% of its neurons sensitive to brain temperature change. It is still unknown whether and how these temperature-sensitive neurons are involved in thermoregulation, because for eight decades they have only been identified via electrophysiological recording. By combining single-cell RNA-seq with whole-cell patch-clamp recordings, we identified Ptgds as a genetic marker for temperature-sensitive POA neurons. Then, we demonstrated these neurons' role in thermoregulation via chemogenetics. Given that Ptgds encodes the enzyme that synthesizes prostaglandin D2 (PGD2), we further explored its role in thermoregulation. Our study revealed that rising temperature of POA alters the activity of Ptgds-expressing neurons so as to increase PGD2 production. PGD2 activates its receptor DP1 and excites downstream neurons in the ventral medial preoptic area (vMPO) that mediates body temperature decrease, a negative feedback loop for thermoregulation.
Collapse
Affiliation(s)
- Tongfei A Wang
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chin Fen Teo
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Malin Åkerblom
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chao Chen
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Marena Tynan-La Fontaine
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Vanille Juliette Greiner
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Aaron Diaz
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Michael T McManus
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh Nung Jan
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Y Jan
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
29
|
Picken C, Fragkos KC, Eddama M, Coghlan G, Clapp LH. Adverse Events of Prostacyclin Mimetics in Pulmonary Arterial Hypertension: A Systematic Review and Meta-Analysis. J Clin Med 2019; 8:E481. [PMID: 30970653 PMCID: PMC6517977 DOI: 10.3390/jcm8040481] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/12/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023] Open
Abstract
Prostacyclin mimetics (PMs) are effective for the treatment of pulmonary arterial hypertension (PAH). However, their clinical use may be limited by their adverse events. This study aims to quantify the different PM adverse events (AEs) with regard to their selectivity towards the prostacyclin (IP) receptor and their administrative routes. The study included randomised, placebo-controlled trials comparing iloprost, beraprost, treprostinil, and selexipag to placebo (published 2002–2016). We report the group efficacy differences between treatment and placebo by weighted and standardised mean difference. The probability of adverse events was determined by the odds ratio (OR). Of the 14 randomised clinical trials involving 3518 PAH patients, outcome and adverse event data were meta-analysed by drug type and route of administration. Prostacyclin mimetics comparison demonstrated a more significant discontinuation of the IP-selective agonist, selexipag, due to an adverse event (OR = 2.2; 95% CI: 1.5, 3.3). Compared to placebo, site pain associated with subcutaneously administered treprostinil was the most significant likely adverse event (OR = 17.5; 95% CI: 11.1, 27.1). Parenteral PMs were associated with fewer adverse effects overall. The overall efficacy of PMs to improve 6-minute walk distance by 16.3 meters was significant (95% CI: 13.0, 19.7). Decreases in pulmonary vascular resistance index (SMD = -5.5; 95% CI: -10.1, -0.9; I² = 98%) and mean pulmonary arterial pressure (SMD = -1.0; 95% CI: -2.6, -0.7; I² = 99%) in treatment groups were found to be significant. Adverse event profiles varied in response to administration route and PM type but were not negated by use of a selective IP agonist. Prostacyclin mimetics exposure to non-target IP receptors may underpin some AEs reported.
Collapse
Affiliation(s)
- Christina Picken
- Institute of Cardiovascular Sciences, University College London, 5 University Street, London, WC1E 6JF, UK.
| | | | - Mohammad Eddama
- Division of Surgery and Interventional Sciences, University College London, 21 University Street, London, WC1E 6AU, UK.
| | - Gerry Coghlan
- Department of Cardiology, Royal Free hospital, London, NW3 2QG, UK.
| | - Lucie H Clapp
- Institute of Cardiovascular Sciences, University College London, 5 University Street, London, WC1E 6JF, UK.
| |
Collapse
|
30
|
Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: Parallels With Inflammatory Processes. Endocr Rev 2019; 40:369-416. [PMID: 30496379 PMCID: PMC6405411 DOI: 10.1210/er.2018-00075] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/18/2018] [Indexed: 12/14/2022]
Abstract
The midcycle surge of LH sets in motion interconnected networks of signaling cascades to bring about rupture of the follicle and release of the oocyte during ovulation. Many mediators of these LH-induced signaling cascades are associated with inflammation, leading to the postulate that ovulation is similar to an inflammatory response. First responders to the LH surge are granulosa and theca cells, which produce steroids, prostaglandins, chemokines, and cytokines, which are also mediators of inflammatory processes. These mediators, in turn, activate both nonimmune ovarian cells as well as resident immune cells within the ovary; additional immune cells are also attracted to the ovary. Collectively, these cells regulate proteolytic pathways to reorganize the follicular stroma, disrupt the granulosa cell basal lamina, and facilitate invasion of vascular endothelial cells. LH-induced mediators initiate cumulus expansion and cumulus oocyte complex detachment, whereas the follicular apex undergoes extensive extracellular matrix remodeling and a loss of the surface epithelium. The remainder of the follicle undergoes rapid angiogenesis and functional differentiation of granulosa and theca cells. Ultimately, these functional and structural changes culminate in follicular rupture and oocyte release. Throughout the ovulatory process, the importance of inflammatory responses is highlighted by the commonalities and similarities between many of these events associated with ovulation and inflammation. However, ovulation includes processes that are distinct from inflammation, such as regulation of steroid action, oocyte maturation, and the eventual release of the oocyte. This review focuses on the commonalities between inflammatory responses and the process of ovulation.
Collapse
Affiliation(s)
- Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - CheMyong Ko
- Department of Comparative Biosciences, University of Illinois Urbana Champaign, Urbana, Illinois
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| | - Mats Brannstrom
- Department of Obstetrics and Gynecology, University of Gothenburg, Gothenburg, Sweden.,Stockholm IVF, Stockholm, Sweden
| | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
31
|
Sonnweber T, Pizzini A, Nairz M, Weiss G, Tancevski I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2018; 19:ijms19113285. [PMID: 30360467 PMCID: PMC6274989 DOI: 10.3390/ijms19113285] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/20/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Lipid and immune pathways are crucial in the pathophysiology of metabolic and cardiovascular disease. Arachidonic acid (AA) and its derivatives link nutrient metabolism to immunity and inflammation, thus holding a key role in the emergence and progression of frequent diseases such as obesity, diabetes, non-alcoholic fatty liver disease, and cardiovascular disease. We herein present a synopsis of AA metabolism in human health, tissue homeostasis, and immunity, and explore the role of the AA metabolome in diverse pathophysiological conditions and diseases.
Collapse
Affiliation(s)
- Thomas Sonnweber
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
32
|
Hanna VS, Hafez EAA. Synopsis of arachidonic acid metabolism: A review. J Adv Res 2018; 11:23-32. [PMID: 30034873 PMCID: PMC6052663 DOI: 10.1016/j.jare.2018.03.005] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/08/2018] [Accepted: 03/11/2018] [Indexed: 12/11/2022] Open
Abstract
Arachidonic acid (AA), a 20 carbon chain polyunsaturated fatty acid with 4 double bonds, is an integral constituent of biological cell membrane, conferring it with fluidity and flexibility. The four double bonds of AA predispose it to oxygenation that leads to a plethora of metabolites of considerable importance for the proper function of the immune system, promotion of allergies and inflammation, resolving of inflammation, mood, and appetite. The present review presents an illustrated synopsis of AA metabolism, corroborating the instrumental importance of AA derivatives for health and well-being. It provides a comprehensive outline on AA metabolic pathways, enzymes and signaling cascades, in order to develop new perspectives in disease treatment and diagnosis.
Collapse
Affiliation(s)
- Violette Said Hanna
- Chemistry Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | | |
Collapse
|
33
|
Kumar S, Palaia T, Hall C, Ragolia L. DP1 receptor agonist, BW245C inhibits diet-induced obesity in ApoE -/- mice. Obes Res Clin Pract 2018; 12:229-241. [PMID: 28602634 DOI: 10.1016/j.orcp.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND/OBJECTIVE Lipocalin Prostaglandin D2 synthase (LPGDS) contributes to the production of PGD2, which has been associated with adipogenesis. In this study, we aimed to investigate the role of PGD2 on obesity through its DP1 and DP2 receptor signaling using intraperitoneal injection of their respective agonists and antagonists. METHODS ApoE-/- mice were divided into five groups: vehicle control (n=5), DP1 receptor agonist (n=5), DP1 receptor antagonist (n=5), DP2 receptor agonist (n=5), and DP2 receptor antagonist (n=5), and the study was carried out for 10 weeks. RESULTS Despite being on high fat diet, mice receiving DP1 receptor agonist sustained a significant inhibition of weight gain throughout the study gaining only 11.4% body weight compared to the controls gaining 61% body weight. Interestingly, parallel to the body weight, the DP1 receptor agonist group showed a significant reduction in food intake throughout the study. Consistently, fasting leptin, insulin and bile acids levels were elevated in the DP1 receptor agonist group compared to controls. As expected, there was a significant reduction in fasting glucose level in DP1 receptor agonist group. At last, as a result of weight gain inhibition, DP1 receptor agonist also imparted cardiovascular benefits showing significant reduction in aortic wall thickness, intima, adventia and lumen size. CONCLUSION Based on the obtained results, we believe DP1 receptor agonism inhibited diet induced weight gain possibly through controlling appetite which consequently imparted beneficial cardiometabolic effects. DP1 receptor agonism may represent a novel therapeutic target for the management of obesity.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Thomas Palaia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Christopher Hall
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Louis Ragolia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States; Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, United States.
| |
Collapse
|
34
|
Pellefigues C, Dema B, Lamri Y, Saidoune F, Chavarot N, Lohéac C, Pacreau E, Dussiot M, Bidault C, Marquet F, Jablonski M, Chemouny JM, Jouan F, Dossier A, Chauveheid MP, Gobert D, Papo T, Karasuyama H, Sacré K, Daugas E, Charles N. Prostaglandin D 2 amplifies lupus disease through basophil accumulation in lymphoid organs. Nat Commun 2018; 9:725. [PMID: 29463843 PMCID: PMC5820278 DOI: 10.1038/s41467-018-03129-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/22/2018] [Indexed: 01/30/2023] Open
Abstract
In systemic lupus erythematosus (SLE), autoantibody production can lead to kidney damage and failure, known as lupus nephritis. Basophils amplify the synthesis of autoantibodies by accumulating in secondary lymphoid organs. Here, we show a role for prostaglandin D2 (PGD2) in the pathophysiology of SLE. Patients with SLE have increased expression of PGD2 receptors (PTGDR) on blood basophils and increased concentration of PGD2 metabolites in plasma. Through an autocrine mechanism dependent on both PTGDRs, PGD2 induces the externalization of CXCR4 on basophils, both in humans and mice, driving accumulation in secondary lymphoid organs. Although PGD2 can accelerate basophil-dependent disease, antagonizing PTGDRs in mice reduces lupus-like disease in spontaneous and induced mouse models. Our study identifies the PGD2/PTGDR axis as a ready-to-use therapeutic modality in SLE.
Collapse
MESH Headings
- Adult
- Animals
- Basophils/immunology
- Female
- Humans
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/immunology
- Lymphatic System/immunology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Prostaglandin D2/blood
- Prostaglandin D2/immunology
- Receptors, CXCR4/blood
- Receptors, CXCR4/immunology
- Receptors, Immunologic/blood
- Receptors, Immunologic/immunology
- Receptors, Prostaglandin/blood
- Receptors, Prostaglandin/immunology
- Signal Transduction/immunology
- Young Adult
Collapse
Affiliation(s)
- Christophe Pellefigues
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Barbara Dema
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Yasmine Lamri
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Fanny Saidoune
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Nathalie Chavarot
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Charlotte Lohéac
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Emeline Pacreau
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Michael Dussiot
- INSERM UMR 1163, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, Institut Imagine, 24 boulevard du Montparnasse, 75015, Paris, France
| | - Caroline Bidault
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Florian Marquet
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
| | - Mathieu Jablonski
- Department of Nephrology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Jonathan M Chemouny
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
- Department of Nephrology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Fanny Jouan
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
- Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Antoine Dossier
- Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Marie-Paule Chauveheid
- Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Delphine Gobert
- Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Thomas Papo
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
- Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Hajime Karasuyama
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
| | - Karim Sacré
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
- Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Eric Daugas
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France
- Department of Nephrology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Faculté de Médecine site Bichat, DHU FIRE, Université Paris Diderot, 46 rue Henri Huchard, 75018, Paris, France
| | - Nicolas Charles
- Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Sorbonne Paris Cité, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, DHU FIRE, Université Paris Diderot, 16 rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
35
|
Li J, Kong D, Wang Q, Wu W, Tang Y, Bai T, Guo L, Wei L, Zhang Q, Yu Y, Qian Y, Zuo S, Liu G, Liu Q, Wu S, Zang Y, Zhu Q, Jia D, Wang Y, Yao W, Ji Y, Yin H, Nakamura M, Lazarus M, Breyer RM, Wang L, Yu Y. Niacin ameliorates ulcerative colitis via prostaglandin D 2-mediated D prostanoid receptor 1 activation. EMBO Mol Med 2017; 9:571-588. [PMID: 28341703 PMCID: PMC5412792 DOI: 10.15252/emmm.201606987] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Niacin, as an antidyslipidemic drug, elicits a strong flushing response by release of prostaglandin (PG) D2. However, whether niacin is beneficial for inflammatory bowel disease (IBD) remains unclear. Here, we observed niacin administration‐enhanced PGD2 production in colon tissues in dextran sulfate sodium (DSS)‐challenged mice, and protected mice against DSS or 2,4,6‐trinitrobenzene sulfonic acid (TNBS)‐induced colitis in D prostanoid receptor 1 (DP1)‐dependent manner. Specific ablation of DP1 receptor in vascular endothelial cells, colonic epithelium, and myeloid cells augmented DSS/TNBS‐induced colitis in mice through increasing vascular permeability, promoting apoptosis of epithelial cells, and stimulating pro‐inflammatory cytokine secretion of macrophages, respectively. Niacin treatment improved vascular permeability, reduced apoptotic epithelial cells, promoted epithelial cell update, and suppressed pro‐inflammatory gene expression of macrophages. Moreover, treatment with niacin‐containing retention enema effectively promoted UC clinical remission and mucosal healing in patients with moderately active disease. Therefore, niacin displayed multiple beneficial effects on DSS/TNBS‐induced colitis in mice by activation of PGD2/DP1 axis. The potential efficacy of niacin in management of IBD warrants further investigation.
Collapse
Affiliation(s)
- Juanjuan Li
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Deping Kong
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qi Wang
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wu
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanping Tang
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Bai
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Guo
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lumin Wei
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qianqian Zhang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu Yu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuting Qian
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengkai Zuo
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guizhu Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qian Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Sheng Wu
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zang
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Zhu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Daile Jia
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyang Wang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Ji
- The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing Jiangsu, China
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Masataka Nakamura
- Human Gene Sciences Center, Tokyo Medical and Dental University, Bunkyo-ku Tokyo, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba City Ibaraki, Japan
| | - Richard M Breyer
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lifu Wang
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Yu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China .,Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
36
|
Ahmad AS, Mendes M, Hernandez D, Doré S. Efficacy of Laropiprant in Minimizing Brain Injury Following Experimental Intracerebral Hemorrhage. Sci Rep 2017; 7:9489. [PMID: 28842638 PMCID: PMC5573370 DOI: 10.1038/s41598-017-09994-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/01/2017] [Indexed: 12/31/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most devastating and disabling forms of stroke, yet effective treatments are still lacking. Prostaglandins and their receptors have been implicated in playing vital roles in ICH outcomes. Recently, laropiprant, a DP1 receptor antagonist, has been used in combination with niacin to abolish the prostaglandin D2-(PGD2)-induced flushing. Here, we test the hypothesis that laropiprant limits bleeding and rescues the brain from ICH. Wildtype (WT) and DP1-/- mice were subjected ICH and neurologic deficits and hemorrhagic lesion outcomes were evaluated at 72 hours after the ICH. To test the therapeutic potential of laropiprant, WT mice subjected to ICH were treated with laropiprant at 1 hour after the ICH. The putative effect of laropiprant on limiting hematoma expansion was tested by an in vivo tail bleeding cessation method and an ex vivo coagulation method. Finally, the roles of laropiprant on gliosis and iron accumulation were also investigated. A significant decrease in the injury volume was observed in DP1-/- as well as laropiprant-treated WT mice. The tail bleeding time was significantly lower in laropiprant group as compared with the vehicle group. Significantly lower Iba-1 and Perls' iron staining in DP1-/- and laropiprant-treated WT groups were observed. Altogether, the data suggest that laropiprant treatment post-ICH attenuates brain damage by targeting primary as well as secondary injuries.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| | - Monique Mendes
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Damian Hernandez
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Departments of Neurology, Psychiatry, Pharmaceutics, Psychology, and Neuroscience, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
37
|
Sandham DA, Barker L, Brown L, Brown Z, Budd D, Charlton SJ, Chatterjee D, Cox B, Dubois G, Duggan N, Hall E, Hatto J, Maas J, Manini J, Profit R, Riddy D, Ritchie C, Sohal B, Shaw D, Stringer R, Sykes DA, Thomas M, Turner KL, Watson SJ, West R, Willard E, Williams G, Willis J. Discovery of Fevipiprant (NVP-QAW039), a Potent and Selective DP 2 Receptor Antagonist for Treatment of Asthma. ACS Med Chem Lett 2017; 8:582-586. [PMID: 28523115 DOI: 10.1021/acsmedchemlett.7b00157] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/25/2017] [Indexed: 12/15/2022] Open
Abstract
Further optimization of an initial DP2 receptor antagonist clinical candidate NVP-QAV680 led to the discovery of a follow-up molecule 2-(2-methyl-1-(4-(methylsulfonyl)-2-(trifluoromethyl)benzyl)-1H-pyrrolo[2,3-b]pyridin-3-yl)acetic acid (compound 11, NVP-QAW039, fevipiprant), which exhibits improved potency on human eosinophils and Th2 cells, together with a longer receptor residence time, and is currently in clinical trials for severe asthma.
Collapse
Affiliation(s)
- David A. Sandham
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Lucy Barker
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Lyndon Brown
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Zarin Brown
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - David Budd
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Steven J. Charlton
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Devnandan Chatterjee
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Brian Cox
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Gerald Dubois
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Nicholas Duggan
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Edward Hall
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Julia Hatto
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Janet Maas
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Jodie Manini
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Rachael Profit
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Darren Riddy
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Catherine Ritchie
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Bindi Sohal
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Duncan Shaw
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Rowan Stringer
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - David A. Sykes
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Matthew Thomas
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Katharine L. Turner
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Simon J. Watson
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Ryan West
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Elisabeth Willard
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Gareth Williams
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| | - Jennifer Willis
- Novartis Institutes for Biomedical Research, Horsham Research Centre, Wimblehurst Road, Horsham, West Sussex RH12 5AB, United Kingdom
| |
Collapse
|
38
|
Kong D, Li J, Shen Y, Liu G, Zuo S, Tao B, Ji Y, Lu A, Lazarus M, Breyer RM, Yu Y. Niacin Promotes Cardiac Healing after Myocardial Infarction through Activation of the Myeloid Prostaglandin D 2 Receptor Subtype 1. J Pharmacol Exp Ther 2017; 360:435-444. [PMID: 28057839 PMCID: PMC5325076 DOI: 10.1124/jpet.116.238261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/30/2016] [Indexed: 01/05/2023] Open
Abstract
Niacin is a well established drug used to lower cholesterol and prevent cardiovascular disease events. However, niacin also causes cutaneous flushing side effects due to release of the proresolution mediator prostaglandin D2 (PGD2). Recent randomized clinical trials have demonstrated that addition of niacin with laropiprant [a PGD2 receptor subtype 1 (DP1) blocker] to statin-based therapies does not significantly decrease the risk of cardiovascular disease events, but increases the risk of serious adverse events. Here, we tested whether, and how, niacin beneficial effects on myocardial ischemia require the activation of the PGD2/DP1 axis. Myocardial infarction (MI) was reproduced by ligation of the left anterior descending branch of the coronary artery in mice. We found that niacin increased PGD2 release in macrophages and shifted macrophages to M2 polarization both in vitro and in vivo by activation of DP1 and accelerated inflammation resolution in zymosan-induced peritonitis in mice. Moreover, niacin treatment facilitated wound healing and improved cardiac function after MI through DP1-mediated M2 bias and timely resolution of inflammation in infarcted hearts. In addition, we found that niacin intake also stimulated M2 polarization of peripheral monocytes in humans. Collectively, niacin promoted cardiac functional recovery after ischemic myocardial infarction through DP1-mediated M2 polarization and timely resolution of inflammation in hearts. These results indicated that DP1 inhibition may attenuate the cardiovascular benefits of niacin.
Collapse
Affiliation(s)
- Deping Kong
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Juanjuan Li
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Yujun Shen
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Guizhu Liu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Shengkai Zuo
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Bo Tao
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Yong Ji
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Ankang Lu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Michael Lazarus
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Richard M Breyer
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (D.K., Y.S., Y.Y.); Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Shanghai, China (D.K., G.L., S.Z., B.T., Y.Y.); Department of Gastroenterology (J.L.), and Department of Cardiology (A.L.); Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; The Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu, China (Y.J.); International Institute for Integrative, Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan (M.L.); and Department of Veterans Affairs, Tennessee Valley Health Authority, and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (R.M.B.)
| |
Collapse
|
39
|
Offermanns S. Hydroxy-Carboxylic Acid Receptor Actions in Metabolism. Trends Endocrinol Metab 2017; 28:227-236. [PMID: 28087125 DOI: 10.1016/j.tem.2016.11.007] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 12/09/2022]
Abstract
Lactic acid, the ketone body 3-hydroxy-butyric acid, also known as β-hydroxybutyrate, and the β-oxidation intermediate 3-hydroxy-octanoic acid are hydroxy-carboxylic acids (HCAs) that serve as intermediates of energy metabolism. However, they also regulate cellular functions, in part by directly activating the G protein-coupled receptors HCA1/GPR81, HCA2/GPR109A, and HCA3/GPR109B. During the past decade, it has become clear that HCA receptors help to maintain homeostasis under changing metabolic and dietary conditions, by controlling metabolic, immune, and other body functions. Work based on genetic mouse models and synthetic ligands of HCA receptors has, in addition, shown that members of this receptor family can serve as targets for the prevention and therapy of diseases such as metabolic and inflammatory disorders.
Collapse
Affiliation(s)
- Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany; Medical Faculty, University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
40
|
Singh U. Efficacy and Safety of Intralesional Xantinol Nicotinate in the Treatment of Various Stages of Oral Submucous Fibrosis. J Clin Diagn Res 2016; 10:ZC34-ZC37. [PMID: 27891454 DOI: 10.7860/jcdr/2016/20138.8639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/07/2016] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Oral Submucous Fibrosis (OSMF) is one of the most prevalent potentially malignant disorders seen in South east population since ages. Despite the extensive amount of research held in this field, its treatment still remains a challenge. In this study, we present our experience in successfully managing OSMF with intralesional injections of a peripheral vasodilator namely xantinol nicotinate. AIM To determine the efficacy and safety of intralesional xantinol nicotinate in the treatment of various stages of OSMF. MATERIALS AND METHODS This parallel, prospective, clinical study included 60 patients clinically diagnosed with oral submucous fibrosis divided into two groups. Group I patients were subjected to intralesional xantinol nicotinate injections bi-weekly for a period of four months while Group II patients were given intralesional saline injections biweekly for four months. All the patients were instructed to perform home mouth opening physiotherapy exercises. At each visit, parameters like increase in interincisal distance, cheek flexibility, tongue protrusion and relief from burning sensation and any side effects were measured and recorded. The drop out figure was zero. RESULTS At the end of four months, in Group I, there was an increase in mean values of interincisal distance, cheek flexibility and tongue protrusion (p<0.001). For burning sensation a significant decrease in mean value was observed (p<0.001). Whereas, in Group 2 the difference between pre-treatment and post-treatment values was not statistically significant (p>0.001). On comparing the results of Group 1 and Group 2, statistically significant difference was observed (p<0.001). CONCLUSION Xantinol nicotinate, a peripheral vasodilator, when injected intralesionally in OSMF patients not only provides relief from burning sensation but also results in increased mouth opening, tongue protrusion and cheek flexibility.
Collapse
Affiliation(s)
- Udita Singh
- Senior Lecturer, Department of Oral Medicine and Radiology, Kothiwal Dental College and Research Centre , Moradabad, Uttar Pradesh, India
| |
Collapse
|
41
|
Effect of PGD 2 on middle meningeal artery and mRNA expression profile of L-PGD 2 synthase and DP receptors in trigeminovascular system and other pain processing structures in rat brain. Pharmacol Rep 2016; 69:50-56. [PMID: 27898338 DOI: 10.1016/j.pharep.2016.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/15/2016] [Accepted: 09/17/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND Prostaglandins (PGs), particularly prostaglandin D2 (PGD2), E2 (PGE2), and I2 (PGI2), are considered to play a role in migraine pain. In humans, infusion of PGD2 causes lesser headache as compared to infusion of PGE2 and PGI2. Follow-up studies in rats have shown that infusion of PGE2 and PGI2 dilate the middle meningeal artery (MMA), and mRNA for PGE2 and PGI2 receptors is present in rat trigeminovascular system (TVS) and in the brain structures associated with pain. In the present study, we have characterized the dilatory effect of PGD2 on rat MMA and studied the relative mRNA expression of PGD2 receptors and lipocalin-type of PGD2 synthase (L-PGDS). METHOD Rat closed-cranial window (CCW) model was used to study the effect of the DP1 receptor antagonist, MK-0524, on PGD2-induced vasodilation of middle meningeal artery. The qPCR technique was used for mRNA expression analysis. RESULTS PGD2 infusion evoked a dose-dependent dilation of the rat MMA. The calculated mean pED50 value was 5.23±0.10 and Emax was 103±18% (n=5). MK-0524 significantly (∼61%, p<0.05) blocked the PGD2-induced dilation of MMA. mRNA for the DP1, DP2 and L-PGDS were expressed differentially in all tested tissues. DP1 receptor mRNA was expressed maximally in trigeminal ganglion (TG) and in cervical dorsal root ganglion (DRG). CONCLUSIONS High expression of DP1 mRNA in the TG and DRG suggest that PGD2 might play a role in migraine pathophysiology. Activation of the DP1 receptor in MMA was mainly responsible for vasodilation induced by PGD2 infusion.
Collapse
|
42
|
The oral CRTh2 antagonist QAW039 (fevipiprant): A phase II study in uncontrolled allergic asthma. Pulm Pharmacol Ther 2016; 39:54-63. [DOI: 10.1016/j.pupt.2016.06.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 06/16/2016] [Accepted: 06/19/2016] [Indexed: 12/20/2022]
|
43
|
Nagira Y, Goto K, Tanaka H, Aoki M, Furue S, Inagaki N, Tomita Y, Shichijo M. Prostaglandin D2 Modulates Neuronal Excitation of the Trigeminal Ganglion to Augment Allergic Rhinitis in Guinea Pigs. J Pharmacol Exp Ther 2016; 357:273-80. [PMID: 26945085 DOI: 10.1124/jpet.115.231225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/02/2016] [Indexed: 03/08/2025] Open
Abstract
Prostaglandin D2(PGD2) is involved in the pathogenesis of allergic rhinitis. However, the sensory nervous system-mediated contributions of PGD2to the symptoms of allergic rhinitis remain unclear. We investigated the involvement of PGD2in these symptoms and in neuronal excitation by in vivo and ex vivo experiments. In an ovalbumin-induced model of allergic rhinitis in guinea pigs, the number of sneezing, nasal rubbing, and nasal secretion events were assessed after the nasal cavity instillation of PGD2, histamine, or a combination of PGD2and histamine. In situ hybridization for PGD2receptor 1 (DP1) mRNA transcripts and immunohistochemical analysis of histamine H1receptor protein expression in guinea pig trigeminal ganglion (TRG) were performed. The effects of DP1receptor activation on the excitability of TRG neurons to electrical and histamine stimuli were assessed using whole-cell patch-clamp recordings. Histamine induced more sneezing, nasal rubbing, and nasal secretion events than PGD2 PGD2augmented histamine-induced responses, whereas pretreatment with a DP1receptor-selective antagonist completely suppressed PGD2-induced augmentation. DP1receptor mRNA transcripts and H1receptor protein expression could be detected in TRG neurons. Moreover, a DP1receptor agonist caused significant increases in the number of histamine-induced action potentials and depolarization, and reduced the current threshold in small-diameter neurons. Our findings show that PGD2-DP1receptor signaling augments the symptoms of allergic rhinitis via the sensory nervous system by modulating nasal neuronal activation to various stimuli, such as histamine. These findings suggest that DP1receptor antagonist has therapeutic potential for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Yoji Nagira
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Kumiko Goto
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Hiroyuki Tanaka
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Miwa Aoki
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Shingo Furue
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Naoki Inagaki
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Yasuhiko Tomita
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| | - Michitaka Shichijo
- United Graduate School of Drug Discovery and Medical Information Sciences (Y.N., H.T., N.I.), Gifu University, and Laboratory of Pharmacology, Department of Bioactive Molecules (H.T., N.I.), Gifu Pharmaceutical University, Gifu, Japan; Discovery Research Laboratory for Innovative Frontier Medicines (Y.N., K.G., S.F., Y.T., M.S.) and Discovery Research Laboratory for Core Therapeutic Areas (M.A.), Shionogi & Co., Ltd., Toyonaka, Osaka, Japan
| |
Collapse
|
44
|
Erpenbeck VJ, Vets E, Gheyle L, Osuntokun W, Larbig M, Neelakantham S, Sandham D, Dubois G, Elbast W, Goldsmith P, Weiss M. Pharmacokinetics, Safety, and Tolerability of Fevipiprant (QAW039), a Novel CRTh2 Receptor Antagonist: Results From 2 Randomized, Phase 1, Placebo-Controlled Studies in Healthy Volunteers. Clin Pharmacol Drug Dev 2016; 5:306-13. [PMID: 27310331 PMCID: PMC5071756 DOI: 10.1002/cpdd.244] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 11/19/2015] [Accepted: 12/01/2015] [Indexed: 12/22/2022]
Abstract
We evaluated the pharmacokinetics (PK), safety, and tolerability of a novel oral CRTh2 antagonist, fevipiprant (QAW039), in healthy subjects. Peak concentrations of fevipiprant in plasma were observed 1‒3 hours postdosing. Concentrations declined in a multiexponential manner, followed by an apparent terminal phase (t1/2, ∼20 hours). Steady state was achieved in 4 days with <2‐fold accumulation. Elimination was partly by renal excretion (≤30% of the dose) and glucuronidation. Food had minimal impact on the PK of fevipiprant, and it was well tolerated at single and multiple oral doses up to 500 mg/day. No dose‐dependent adverse events were observed, and all the events were mild or moderate in severity. Systemic concentrations were sufficiently high to achieve relevant target occupancy, considering in vitro pharmacology data. In summary, the data support further development as a once‐daily oral therapy for allergic diseases.
Collapse
Affiliation(s)
| | - Eva Vets
- SGS-Life Sciences, Antwerp, Belgium
| | | | - Wande Osuntokun
- Novartis Institutes for Biomedical Research, Horsham, United Kingdom
| | | | | | - David Sandham
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Gerald Dubois
- Novartis Institutes for Biomedical Research, Horsham, United Kingdom
| | | | - Paul Goldsmith
- Novartis Institutes for Biomedical Research, Horsham, United Kingdom
| | | |
Collapse
|
45
|
Abstract
Niacin is effective in treating dyslipidemias but causes cutaneous vasodilation or flushing, a side effect that limits its clinical use. Blocking prostaglandins in humans reduces but does not consistently eliminate flushing, indicating additional mechanisms may contribute to flushing. The transient receptor potential vanilloid 1 (TRPV1) channel, when activated, causes cutaneous vasodilation and undergoes tachyphylaxis similar to that seen with niacin. Using a murine model, early phase niacin-induced flushing was examined and TRPV1 channel involvement demonstrated using pharmacologic blockade, desensitization, and genetic knockouts (TRPV1 KO). The TRPV1 antagonist AMG9810 reduced the magnitude of the initial and secondary peaks and the rapidity of the vasodilatory response (slope). TRPV1 desensitization by chronic capsaicin reduced the initial peak and slope. TRPV1 KO mice had a lower initial peak, secondary peak, and slope compared with wild-type mice. Chronic niacin reduced the initial peak, secondary peak, and slope in wild-type mice but had no effect in knockout mice. Furthermore, chronic niacin diminished the response to capsaicin in wild-type mice. Overall, these data demonstrate an important role for TRPV1 channels in niacin-induced flushing, both in the acute response and with chronic administration. That niacin-induced flushing is a complex cascade of events, which should inform pharmacological intervention against this side effect.
Collapse
|
46
|
Banipal TS, Kaur A, Khan IA, Banipal PK. Exploring the thermodynamics and conformational aspects of nicotinic acid binding with bovine serum albumin: a detailed calorimetric, spectroscopic and molecular docking study. RSC Adv 2016. [DOI: 10.1039/c5ra28028a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
An attempt to obtain a physicochemical and conformational outlook on the binding interaction of vitamin B3 (NA) with a model transport protein BSA using calorimetry, light scattering, molecular docking, and spectroscopic techniques.
Collapse
Affiliation(s)
| | - Amandeep Kaur
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar – 143005
- India
| | - Imran Ahmd Khan
- Department of Chemistry
- Guru Nanak Dev University
- Amritsar – 143005
- India
| | | |
Collapse
|
47
|
McNamara MJ, Sayanlar JJ, Dooley DJ, Srichai MB, Taylor AJ. A randomized pilot study on the effect of niacin on pulmonary arterial pressure. Trials 2015; 16:530. [PMID: 26590128 PMCID: PMC4654874 DOI: 10.1186/s13063-015-1013-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 10/15/2015] [Indexed: 11/11/2022] Open
Abstract
Background Niacin induces the release of vasodilating prostaglandins, for which receptors are present within the pulmonary arterial circulation. We hypothesized that immediate-release niacin would reduce right ventricular systolic pressure in patients with pulmonary hypertension in a randomized, double-blinded, single-dose provocation study. Methods We recruited inpatient subjects with a Doppler echocardiogram showing a peak tricuspid regurgitation (TR) jet velocity of 2.7 m/s or greater, and who were free of known pulmonary vascular disease. Subjects were randomized in a 1:2:2 ratio to receive a single dose of either placebo, niacin 100 mg or niacin 500 mg, respectively. TR jet velocities were measured immediately before, and 1 hour post dose, corresponding to peak niacin absorption and prostaglandin release. The primary endpoint was the change in mean TR jet velocity measured over ten successive cardiac cycles. Results The baseline mean estimated right ventricular systolic pressure (RVSP) for all 49 subjects (25 male) was 51.9 ± 12.1 mm Hg. The primary endpoint of mean change in TR jet velocity was 0.016 ± 0.065 m/s in the placebo group, compared to −0.017 ± 0.065 m/s with niacin 100 mg, and −0.063 ± 0.038 m/s with niacin 500 mg (P = 0.63). The change in maximum estimated RVSP across the three drug groups was 0.2 ± 1.6 mm Hg, −1.3 ± 1.8 mm Hg and −2.2 ± 1.2 mm Hg (P = 0.62). In exploratory pairwise analysis in the high-dose niacin group (500 mg), the reduction in mean RVSP was from 50.9 ± 9.4 mm Hg to 48.7 ± 10.0 mm Hg (P = 0.09). Conclusions A single dose of immediate-release niacin (100 mg or 500 mg) had no significant effect on RVSP 1 hour post administration. A nonsignificant dose-dependent trend for a modest reduction in RVSP, most notable in the 500 mg group, was noted. (ISRCTN number 12353191, registered April 23, 2015). Electronic supplementary material The online version of this article (doi:10.1186/s13063-015-1013-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin J McNamara
- Cardiology Division, Medstar Heart and Vascular Institute at Medstar Georgetown University Hospital and Medstar Washington Hospital Center, 3800 Reservoir Rd., NW, 5 PHC, Washington, DC, 20007, USA.
| | - Jason J Sayanlar
- Cardiology Division, Medstar Heart and Vascular Institute at Medstar Georgetown University Hospital and Medstar Washington Hospital Center, 3800 Reservoir Rd., NW, 5 PHC, Washington, DC, 20007, USA.
| | - Daniel J Dooley
- Cardiology Division, Medstar Heart and Vascular Institute at Medstar Georgetown University Hospital and Medstar Washington Hospital Center, 3800 Reservoir Rd., NW, 5 PHC, Washington, DC, 20007, USA.
| | - Monvadi B Srichai
- Cardiology Division, Medstar Heart and Vascular Institute at Medstar Georgetown University Hospital and Medstar Washington Hospital Center, 3800 Reservoir Rd., NW, 5 PHC, Washington, DC, 20007, USA.
| | - Allen J Taylor
- Cardiology Division, Medstar Heart and Vascular Institute at Medstar Georgetown University Hospital and Medstar Washington Hospital Center, 3800 Reservoir Rd., NW, 5 PHC, Washington, DC, 20007, USA.
| |
Collapse
|
48
|
Zhou H, Zhu Q, Gan Z, Dong G, Xu Y. Synthesis and biological evaluation of novel laropiprant derivatives as potential anti-allergic agents. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1431-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
49
|
Laidlaw TM, Boyce JA. Platelets in patients with aspirin-exacerbated respiratory disease. J Allergy Clin Immunol 2015; 135:1407-14; quiz 1415. [PMID: 26051947 DOI: 10.1016/j.jaci.2015.02.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/26/2015] [Accepted: 02/04/2015] [Indexed: 12/12/2022]
Abstract
Aspirin-exacerbated respiratory disease (AERD) is a chronic inflammatory disease characterized clinically by the triad of asthma, nasal polyposis, and pathognomonic respiratory reactions after ingestion of aspirin. It is a distinct syndrome associated with eosinophilic infiltration of respiratory tissues and excessive production of cysteinyl leukotrienes. Despite the consistent clinical phenotype of the respiratory disease, the underlying pathogenesis of the disease remains unclear. In addition to their role in hemostasis, platelets have the capacity to influence the activation state and function of other immune cells during inflammation and to facilitate granulocyte recruitment into the tissues. Platelets also possess a repertoire of potent preformed mediators of inflammation that are released on activation and are a rich source of newly synthesized lipid mediators that alter vascular permeability and smooth muscle tone. Accordingly, platelet activity has been linked to diverse inflammatory diseases, including asthma. Both human and animal studies strongly suggest that platelet activity is uniquely associated with the pathophysiology of AERD. This article summarizes the evidence supporting an effector role for platelets in asthmatic patients in general and in patients with AERD in particular and considers the potential therapeutic implications.
Collapse
Affiliation(s)
- Tanya M Laidlaw
- Department of Medicine, Harvard Medical School, Boston, Mass; Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, Mass.
| | - Joshua A Boyce
- Department of Medicine, Harvard Medical School, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, Mass
| |
Collapse
|
50
|
Abstract
Controlled immune responses to infection and injury involve complex molecular signalling networks with coordinated and often opposing actions. Eicosanoids and related bioactive lipid mediators derived from polyunsaturated fatty acids constitute a major bioactive lipid network that is among the most complex and challenging pathways to map in a physiological context. Eicosanoid signalling, similar to cytokine signalling and inflammasome formation, has primarily been viewed as a pro-inflammatory component of the innate immune response; however, recent advances in lipidomics have helped to elucidate unique eicosanoids and related docosanoids with anti-inflammatory and pro-resolution functions. This has advanced our overall understanding of the inflammatory response and its therapeutic implications. The induction of a pro-inflammatory and anti-inflammatory eicosanoid storm through the activation of inflammatory receptors by infectious agents is reviewed here.
Collapse
Affiliation(s)
- Edward A Dennis
- Department of Chemistry and Biochemistry and Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Paul C Norris
- Department of Chemistry and Biochemistry and Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|